Publications

Professor of Psychiatry and Behavioral Sciences (Major Laboratories and Clinical and Translational Neurosciences Incubator)

Publications

  • Sleep and vigilance states: Embracing spatiotemporal dynamics. Neuron Nir, Y., de Lecea, L. 2023

    Abstract

    The classic view of sleep and vigilance states is a global stationary perspective driven by the interaction betweenneuromodulators and thalamocortical systems. However, recent data are challenging this view by demonstrating that vigilance states are highly dynamic and regionally complex. Spatially, sleep- and wake-like states often co-occur across distinct brain regions, as in unihemispheric sleep, local sleep in wakefulness, and during development. Temporally, dynamic switching prevails around state transitions, during extended wakefulness, and in fragmented sleep. This knowledge, together with methods monitoring brain activity across multiple regions simultaneously at millisecond resolution with cell-type specificity, is rapidly shifting how we consider vigilance states. A new perspective incorporating multiple spatial and temporal scales may have important implications for considering the governing neuromodulatory mechanisms, the functional roles of vigilance states, and their behavioral manifestations. A modular and dynamic view highlights novel avenues for finer spatiotemporal interventions to improve sleep function.

    View details for DOI 10.1016/j.neuron.2023.04.012

    View details for PubMedID 37148873

  • A tool for monitoring cell type-specific focused ultrasound neuromodulation and control of chronic epilepsy. Proceedings of the National Academy of Sciences of the United States of America Murphy, K. R., Farrell, J. S., Gomez, J. L., Stedman, Q. G., Li, N., Leung, S. A., Good, C. H., Qiu, Z., Firouzi, K., Butts Pauly, K., Khuri-Yakub, B. P., Michaelides, M., Soltesz, I., de Lecea, L. 2022; 119 (46): e2206828119

    Abstract

    Focused ultrasound (FUS) is a powerful tool for noninvasive modulation of deep brain activity with promising therapeutic potential for refractory epilepsy; however, tools for examining FUS effects on specific cell types within the deep brain do not yet exist. Consequently, how cell types within heterogeneous networks can be modulated and whether parameters can be identified to bias these networks in the context of complex behaviors remains unknown. To address this, we developed a fiber Photometry Coupled focused Ultrasound System (PhoCUS) for simultaneously monitoring FUS effects on neural activity of subcortical genetically targeted cell types in freely behaving animals. We identified a parameter set that selectively increases activity of parvalbumin interneurons while suppressing excitatory neurons in the hippocampus. A net inhibitory effect localized to the hippocampus was further confirmed through whole brain metabolic imaging. Finally, these inhibitory selective parameters achieved significant spike suppression in the kainate model of chronic temporal lobe epilepsy, opening the door for future noninvasive therapies.

    View details for DOI 10.1073/pnas.2206828119

    View details for PubMedID 36343238

  • Adolescent sleep shapes social novelty preference in mice. Nature neuroscience Bian, W. J., Brewer, C. L., Kauer, J. A., de Lecea, L. 2022

    Abstract

    Sleep disturbances frequently occur in neurodevelopmental disorders such as autism, but the developmental role of sleep is largely unexplored, and a causal relationship between developmental sleep defects and behavioral consequences in adulthood remains elusive. Here, we show that in mice, sleep disruption (SD) in adolescence, but not in adulthood, causes long-lasting impairment in social novelty preference. Furthermore, adolescent SD alters the activation and release patterns of dopaminergic neurons in the ventral tegmental area (VTA) in response to social novelty. This developmental sleep function is mediated by balanced VTA activity during adolescence; chemogenetic excitation mimics, whereas silencing rescues, the social deficits of adolescent SD. Finally, we show that in Shank3-mutant mice, improving sleep or rectifying VTA activity during adolescence ameliorates adult social deficits. Together, our results identify a critical role of sleep and dopaminergic activity in the development of social interaction behavior.

    View details for DOI 10.1038/s41593-022-01076-8

    View details for PubMedID 35618950

  • Hyperexcitable arousal circuits drive sleep instability during aging. Science (New York, N.Y.) Li, S. B., Damonte, V. M., Chen, C., Wang, G. X., Kebschull, J. M., Yamaguchi, H., Bian, W. J., Purmann, C., Pattni, R., Urban, A. E., Mourrain, P., Kauer, J. A., Scherrer, G., de Lecea, L. 2022; 375 (6583): eabh3021

    Abstract

    Sleep quality declines with age; however, the underlying mechanisms remain elusive. We found that hyperexcitable hypocretin/orexin (Hcrt/OX) neurons drive sleep fragmentation during aging. In aged mice, Hcrt neurons exhibited more frequent neuronal activity epochs driving wake bouts, and optogenetic activation of Hcrt neurons elicited more prolonged wakefulness. Aged Hcrt neurons showed hyperexcitability with lower KCNQ2 expression and impaired M-current, mediated by KCNQ2/3 channels. Single-nucleus RNA-sequencing revealed adaptive changes to Hcrt neuron loss in the aging brain. Disruption of Kcnq2/3 genes in Hcrt neurons of young mice destabilized sleep, mimicking aging-associated sleep fragmentation, whereas the KCNQ-selective activator flupirtine hyperpolarized Hcrt neurons and rejuvenated sleep architecture in aged mice. Our findings demonstrate a mechanism underlying sleep instability during aging and a strategy to improve sleep continuity.

    View details for DOI 10.1126/science.abh3021

    View details for PubMedID 35201886

  • Parallel circuits from the bed nuclei of stria terminalis to the lateral hypothalamus drive opposing emotional states NATURE NEUROSCIENCE Giardino, W. J., Eban-Rothschild, A., Christoffel, D. J., Li, S., Malenka, R. C., de Lecea, L. 2018; 21 (8): 1084-+
  • VTA dopaminergic neurons regulate ethologically relevant sleep-wake behaviors. Nature neuroscience Eban-Rothschild, A., Rothschild, G., Giardino, W. J., Jones, J. R., de Lecea, L. 2016; 19 (10): 1356-1366

    Abstract

    Dopaminergic ventral tegmental area (VTA) neurons are critically involved in a variety of behaviors that rely on heightened arousal, but whether they directly and causally control the generation and maintenance of wakefulness is unknown. We recorded calcium activity using fiber photometry in freely behaving mice and found arousal-state-dependent alterations in VTA dopaminergic neurons. We used chemogenetic and optogenetic manipulations together with polysomnographic recordings to demonstrate that VTA dopaminergic neurons are necessary for arousal and that their inhibition suppresses wakefulness, even in the face of ethologically relevant salient stimuli. Nevertheless, before inducing sleep, inhibition of VTA dopaminergic neurons promoted goal-directed and sleep-related nesting behavior. Optogenetic stimulation, in contrast, initiated and maintained wakefulness and suppressed sleep and sleep-related nesting behavior. We further found that different projections of VTA dopaminergic neurons differentially modulate arousal. Collectively, our findings uncover a fundamental role for VTA dopaminergic circuitry in the maintenance of the awake state and ethologically relevant sleep-related behaviors.

    View details for DOI 10.1038/nn.4377

    View details for PubMedID 27595385

  • Tuning arousal with optogenetic modulation of locus coeruleus neurons NATURE NEUROSCIENCE Carter, M. E., Yizhar, O., Chikahisa, S., Nguyen, H., Adamantidis, A., Nishino, S., Deisseroth, K., de Lecea, L. 2010; 13 (12): 1526-U117

    Abstract

    Neural activity in the noradrenergic locus coeruleus correlates with periods of wakefulness and arousal. However, it is unclear whether tonic or phasic activity in these neurons is necessary or sufficient to induce transitions between behavioral states and to promote long-term arousal. Using optogenetic tools in mice, we found that there is a frequency-dependent, causal relationship among locus coeruleus firing, cortical activity, sleep-to-wake transitions and general locomotor arousal. We also found that sustained, high-frequency stimulation of the locus coeruleus at frequencies of 5 Hz and above caused reversible behavioral arrests. These results suggest that the locus coeruleus is finely tuned to regulate organismal arousal and that bursts of noradrenergic overexcitation cause behavioral attacks that resemble those seen in people with neuropsychiatric disorders.

    View details for DOI 10.1038/nn.2682

    View details for Web of Science ID 000284525800018

    View details for PubMedID 21037585

    View details for PubMedCentralID PMC3174240

  • Phasic Firing in Dopaminergic Neurons Is Sufficient for Behavioral Conditioning SCIENCE Tsai, H., Zhang, F., Adamantidis, A., Stuber, G. D., Bonci, A., de Lecea, L., Deisseroth, K. 2009; 324 (5930): 1080-1084

    Abstract

    Natural rewards and drugs of abuse can alter dopamine signaling, and ventral tegmental area (VTA) dopaminergic neurons are known to fire action potentials tonically or phasically under different behavioral conditions. However, without technology to control specific neurons with appropriate temporal precision in freely behaving mammals, the causal role of these action potential patterns in driving behavioral changes has been unclear. We used optogenetic tools to selectively stimulate VTA dopaminergic neuron action potential firing in freely behaving mammals. We found that phasic activation of these neurons was sufficient to drive behavioral conditioning and elicited dopamine transients with magnitudes not achieved by longer, lower-frequency spiking. These results demonstrate that phasic dopaminergic activity is sufficient to mediate mammalian behavioral conditioning.

    View details for DOI 10.1126/science.1168878

    View details for Web of Science ID 000266246700044

    View details for PubMedID 19389999

  • Neural substrates of awakening probed with optogenetic control of hypocretin neurons NATURE Adamantidis, A. R., Zhang, F., Aravanis, A. M., Deisseroth, K., de Lecea, L. 2007; 450 (7168): 420-U9

    Abstract

    The neural underpinnings of sleep involve interactions between sleep-promoting areas such as the anterior hypothalamus, and arousal systems located in the posterior hypothalamus, the basal forebrain and the brainstem. Hypocretin (Hcrt, also known as orexin)-producing neurons in the lateral hypothalamus are important for arousal stability, and loss of Hcrt function has been linked to narcolepsy. However, it is unknown whether electrical activity arising from Hcrt neurons is sufficient to drive awakening from sleep states or is simply correlated with it. Here we directly probed the impact of Hcrt neuron activity on sleep state transitions with in vivo neural photostimulation, genetically targeting channelrhodopsin-2 to Hcrt cells and using an optical fibre to deliver light deep in the brain, directly into the lateral hypothalamus, of freely moving mice. We found that direct, selective, optogenetic photostimulation of Hcrt neurons increased the probability of transition to wakefulness from either slow wave sleep or rapid eye movement sleep. Notably, photostimulation using 5-30 Hz light pulse trains reduced latency to wakefulness, whereas 1 Hz trains did not. This study establishes a causal relationship between frequency-dependent activity of a genetically defined neural cell type and a specific mammalian behaviour central to clinical conditions and neurobehavioural physiology.

    View details for DOI 10.1038/nature06310

    View details for Web of Science ID 000250918600055

    View details for PubMedID 17943086

  • The murine meninges acquire lymphoid tissue properties and harbour autoreactive B cells during chronic Trypanosoma brucei infection. PLoS biology Quintana, J. F., Sinton, M. C., Chandrasegaran, P., Kumar Dubey, L., Ogunsola, J., Al Samman, M., Haley, M., McConnell, G., Kuispond Swar, N. R., Ngoyi, D. M., Bending, D., de Lecea, L., MacLeod, A., Mabbott, N. A. 2023; 21 (11): e3002389

    Abstract

    The meningeal space is a critical brain structure providing immunosurveillance for the central nervous system (CNS), but the impact of infections on the meningeal immune landscape is far from being fully understood. The extracellular protozoan parasite Trypanosoma brucei, which causes human African trypanosomiasis (HAT) or sleeping sickness, accumulates in the meningeal spaces, ultimately inducing severe meningitis and resulting in death if left untreated. Thus, sleeping sickness represents an attractive model to study immunological dynamics in the meninges during infection. Here, by combining single-cell transcriptomics and mass cytometry by time-of-flight (CyTOF) with in vivo interventions, we found that chronic T. brucei infection triggers the development of ectopic lymphoid aggregates (ELAs) in the murine meninges. These infection-induced ELAs were defined by the presence of ER-TR7+ fibroblastic reticular cells, CD21/35+ follicular dendritic cells (FDCs), CXCR5+ PD1+ T follicular helper-like phenotype, GL7+ CD95+ GC-like B cells, and plasmablasts/plasma cells. Furthermore, the B cells found in the infected meninges produced high-affinity autoantibodies able to recognise mouse brain antigens, in a process dependent on LTβ signalling. A mid-throughput screening identified several host factors recognised by these autoantibodies, including myelin basic protein (MBP), coinciding with cortical demyelination and brain pathology. In humans, we identified the presence of autoreactive IgG antibodies in the cerebrospinal fluid (CSF) of second stage HAT patients that recognised human brain lysates and MBP, consistent with our findings in experimental infections. Lastly, we found that the pathological B cell responses we observed in the meninges required the presence of T. brucei in the CNS, as suramin treatment before the onset of the CNS stage prevented the accumulation of GL7+ CD95+ GC-like B cells and brain-specific autoantibody deposition. Taken together, our data provide evidence that the meningeal immune response during chronic T. brucei infection results in the acquisition of lymphoid tissue-like properties, broadening our understanding of meningeal immunity in the context of chronic infections. These findings have wider implications for understanding the mechanisms underlying the formation ELAs during chronic inflammation resulting in autoimmunity in mice and humans, as observed in other autoimmune neurodegenerative disorders, including neuropsychiatric lupus and multiple sclerosis.

    View details for DOI 10.1371/journal.pbio.3002389

    View details for PubMedID 37983289

  • Dorsomedial and preoptic hypothalamic circuits control torpor. Current biology : CB Yamaguchi, H., Murphy, K. R., Fukatsu, N., Sato, K., Yamanaka, A., de Lecea, L. 2023

    Abstract

    Endotherms can survive low temperatures and food shortage by actively entering a hypometabolic state known as torpor. Although the decrease in metabolic rate and body temperature (Tb) during torpor is controlled by the brain, the specific neural circuits underlying these processes have not been comprehensively elucidated. In this study, we identify the neural circuits involved in torpor regulation by combining whole-brain mapping of torpor-activated neurons, cell-type-specific manipulation of neural activity, and viral tracing-based circuit mapping. We find that Trpm2-positive neurons in the preoptic area and Vgat-positive neurons in the dorsal medial hypothalamus are activated during torpor. Genetic silencing shows that the activity of either cell type is necessary to enter the torpor state. Finally, we show that these cells receive projections from the arcuate and suprachiasmatic nucleus and send projections to brain regions involved in thermoregulation. Our results demonstrate an essential role of hypothalamic neurons in the regulation of Tb and metabolic rate during torpor and identify critical nodes of the torpor regulatory network.

    View details for DOI 10.1016/j.cub.2023.10.076

    View details for PubMedID 37992720

  • Sleep and the hypothalamus. Science (New York, N.Y.) Adamantidis, A. R., de Lecea, L. 2023; 382 (6669): 405-412

    Abstract

    Neural substrates of wakefulness, rapid eye movement sleep (REMS), and non-REMS (NREMS) in the mammalian hypothalamus overlap both anatomically and functionally with cellular networks that support physiological and behavioral homeostasis. Here, we review the roles of sleep neurons of the hypothalamus in the homeostatic control of thermoregulation or goal-oriented behaviors during wakefulness. We address how hypothalamic circuits involved in opposing behaviors such as core body temperature and sleep compute conflicting information and provide a coherent vigilance state. Finally, we highlight some of the key unresolved questions and challenges, and the promise of a more granular view of the cellular and molecular diversity underlying the integrative role of the hypothalamus in physiological and behavioral homeostasis.

    View details for DOI 10.1126/science.adh8285

    View details for PubMedID 37883555

  • Cell type specific focused ultrasound neuromodulation in preclinical models of sleep and psychiatric disorders. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology Murphy, K. R., de Lecea, L. 2023

    View details for DOI 10.1038/s41386-023-01662-9

    View details for PubMedID 37463978

  • The Effect of Food Motivational Status and Hypocretin (Orexin) 1 Receptor Antagonism on Decision Processes in the Murine Go/No-Go Behavioural Task Jacobson, L., Ji, M., Metha, J., Murawski, C., Nicholson, J., De Lecea, L., Hoyer, D. WILEY. 2023: 1025-1026
  • Adolescent sleep defects and dopaminergic hyperactivity in mice with a schizophrenia-linked Shank3 mutation. Sleep Bian, W. J., González, O. C., de Lecea, L. 2023

    Abstract

    Shank3 is a shared risk gene for autism spectrum disorders and schizophrenia. Sleep defects have been characterized for autism models with Shank3 mutations, however, evidence has been lacking for the potential sleep defects caused by Shank3 mutation associated with schizophrenia and how early in development these defects may occur. Here we characterized the sleep architecture of adolescent mice carrying a schizophrenia-linked, R1117X mutation in Shank3. We further employed GRABDA dopamine sensor and fiber photometry to record dopamine release in the nucleus accumbens during sleep/wake states. Our results show that homozygous mutant R1117X mice have significantly reduced sleep in the dark phase during adolescence, altered electroencephalogram power especially during the rapid-eye-movement sleep, and dopamine hyperactivity during sleep but not during wakefulness. Further analyses suggest that these adolescent defects in sleep architecture and dopaminergic neuromodulation tightly correlate with the social novelty preference later in adulthood and predict adult social performance during same-sex social interactions. Our results provide novel insights to the sleep phenotypes in mouse models of schizophrenia and the potential use of developmental sleep as a predictive metric for adult social symptoms. Together with recent studies in other Shank3 models, our work underscores the idea that Shank3-involved circuit disruptions may be one of the shared pathologies in certain types of schizophrenia and autism. Future research is needed to establish the causal relationship among adolescent sleep defects, dopaminergic dysregulation, and adult behavioral changes in Shank3 mutation animals and other models.

    View details for DOI 10.1093/sleep/zsad131

    View details for PubMedID 37144901

  • Myeloid deficiency of the intrinsic clock protein BMAL1 accelerates cognitive aging by disrupting microglial synaptic pruning. Journal of neuroinflammation Iweka, C. A., Seigneur, E., Hernandez, A. L., Paredes, S. H., Cabrera, M., Blacher, E., Pasternak, C. T., Longo, F. M., de Lecea, L., Andreasson, K. I. 2023; 20 (1): 48

    Abstract

    Aging is associated with loss of circadian immune responses and circadian gene transcription in peripheral macrophages. Microglia, the resident macrophages of the brain, also show diurnal rhythmicity in regulating local immune responses and synaptic remodeling. To investigate the interaction between aging and microglial circadian rhythmicity, we examined mice deficient in the core clock transcription factor, BMAL1. Aging Cd11bcre;Bmallox/lox mice demonstrated accelerated cognitive decline in association with suppressed hippocampal long-term potentiation and increases in immature dendritic spines. C1q deposition at synapses and synaptic engulfment were significantly decreased in aging Bmal1-deficient microglia, suggesting that BMAL1 plays a role in regulating synaptic pruning in aging. In addition to accelerated age-associated hippocampal deficits, Cd11bcre;Bmallox/lox mice also showed deficits in the sleep-wake cycle with increased wakefulness across light and dark phases. These results highlight an essential role of microglial BMAL1 in maintenance of synapse homeostasis in the aging brain.

    View details for DOI 10.1186/s12974-023-02727-8

    View details for PubMedID 36829230

    View details for PubMedCentralID 8157517

  • Automated Sleep Deprivation Setup Using a Shaking Platform in Mice. Bio-protocol Bian, W., De Lecea, L. 2023; 13 (4): e4620

    Abstract

    The functions of sleep remain largely unclear, and even less is known about its role in development. A general strategy to tackle these questions is to disrupt sleep and measure the outcomes. However, some existing sleep deprivation methods may not be suitable for studying the effects of chronic sleep disruption, due to their lack of effectiveness and/or robustness, substantial stress caused by the deprivation method, or consuming a large quantity of time and manpower. More problems may be encountered when applying these existing protocols to young, developing animals, because of their likely heightened vulnerability to stressors, and difficulties in precisely monitoring sleep at young ages. Here, we report a protocol of automated sleep disruption in mice using a commercially available, shaking platform-based deprivation system. We show that this protocol effectively and robustly deprives both non-rapid-eye-movement (NREM) sleep and rapid-eye-movement (REM) sleep without causing a significant stress response, and does not require human supervision. This protocol uses adolescent mice, but the method also works with adult mice. Graphical abstract Automated sleep deprivation system. The platform of the deprivation chamber was programmed to shake in a given frequency and intensity to keep the animal awake while its brain and muscle activities were continuously monitored by electroencephalography and electromyography.

    View details for DOI 10.21769/BioProtoc.4620

    View details for PubMedID 36845529

  • Optogenetic and pharmacological interventions link hypocretin neurons to impulsivity in mice. Communications biology Tyree, S. M., Jennings, K. J., Gonzalez, O. C., Li, S., Nicholson, J. R., von Heimendahl, M., de Lecea, L. 2023; 6 (1): 74

    Abstract

    Neurons in the lateral hypothalamus expressing the neuropeptide Hypocretin, also known as orexin, are known critical modulators of arousal stability. However, their role in the different components of the arousal construct such as attention and decision making is poorly understood. Here we study Hypocretin neuronal circuit dynamics during stop action impulsivity in a Go/NoGo task in mice. We show that Hypocretin neuronal activity correlates with anticipation of reward. We then assessed the causal role of Hypocretin neuronal activity using optogenetics in a Go/NoGo task. We show that stimulation of Hypocretin neurons during the cue period dramatically increases the number of premature responses. These effects are mimicked by amphetamine, reduced by atomoxetine, a norepinephrine uptake inhibitor, and blocked by a Hypocretin receptor 1 selective antagonist. We conclude that Hypocretin neurons have a key role in the integration of salient stimuli during wakefulness to produce appropriate and timely responses to rewarding and aversive cues.

    View details for DOI 10.1038/s42003-023-04409-w

    View details for PubMedID 36658362

  • Single cell and spatial transcriptomic analyses reveal microglia-plasma cell crosstalk in the brain during Trypanosoma brucei infection. Nature communications Quintana, J. F., Chandrasegaran, P., Sinton, M. C., Briggs, E. M., Otto, T. D., Heslop, R., Bentley-Abbot, C., Loney, C., de Lecea, L., Mabbott, N. A., MacLeod, A. 2022; 13 (1): 5752

    Abstract

    Human African trypanosomiasis, or sleeping sickness, is caused by the protozoan parasite Trypanosoma brucei and induces profound reactivity of glial cells and neuroinflammation when the parasites colonise the central nervous system. However, the transcriptional and functional responses of the brain to chronic T. brucei infection remain poorly understood. By integrating single cell and spatial transcriptomics of the mouse brain, we identify that glial responses triggered by infection are readily detected in the proximity to the circumventricular organs, including the lateral and 3rd ventricle. This coincides with the spatial localisation of both slender and stumpy forms of T. brucei. Furthermore, in silico predictions and functional validations led us to identify a previously unknown crosstalk between homeostatic microglia and Cd138+ plasma cells mediated by IL-10 and B cell activating factor (BAFF) signalling. This study provides important insights and resources to improve understanding of the molecular and cellular responses in the brain during infection with African trypanosomes.

    View details for DOI 10.1038/s41467-022-33542-z

    View details for PubMedID 36180478

  • The brake matters: Hyperexcitable arousal circuits in sleep fragmentation with age. Clinical and translational medicine Li, S., de Lecea, L. 2022; 12 (6): e900

    View details for DOI 10.1002/ctm2.900

    View details for PubMedID 35696605

  • Lateral hypothalamic galanin neurons are activated by stress and blunt anxiety-like behavior in mice. Behavioural brain research Owens-French, J., Li, S., Francois, M., Leigh Townsend, R., Daniel, M., Soulier, H., Turner, A., de Lecea, L., Munzberg, H., Morrison, C., Qualls-Creekmore, E. 1800: 113773

    Abstract

    Despite the prevalence of anxiety disorders, the molecular identity of neural circuits underlying anxiety remains unclear. The lateral hypothalamus (LH) is one brain region implicated in the regulation of anxiety, and our recent data found that chemogenetic activation of LH galanin neurons attenuated the stress response to a novel environment as measured by the marble burying test. Thus, we hypothesize that LH galanin neurons may contribute to anxiety-related behavior. We used chemogenetics and fiber photometry to test the ability of LH galanin neurons to influence anxiety and stress-related behavior. Chemogenetic activation of LH galanin neurons significantly decreased anxiety-like behavior in the elevated plus maze, open field test, and light dark test. However, LH galanin activation did not alter restraint stress induced HPA activation or freezing behavior in the fear conditioning paradigm. In vivo calcium monitoring by fiber photometry indicated that LH galanin neurons were activated by anxiogenic and/or stressful stimuli including tail suspension, novel mouse interaction, and predator odor. Further, in a fear conditioning task, calcium transients strongly increased during foot shock, but were not affected by the unconditioned stimulus tone. These data indicate that LH galanin neurons both respond to and modulate anxiety, with no influence on stress induced HPA activation or fear behaviors. Further investigation of LH galanin circuitry and functional mediators of behavioral output may offer a more refined pharmacological target as an alternative to first-line broad pharmacotherapies such as benzodiazepines.

    View details for DOI 10.1016/j.bbr.2022.113773

    View details for PubMedID 35101456

  • Hypocretins (orexins): The ultimate translational neuropeptides. Journal of internal medicine Jacobson, L. H., Hoyer, D., de Lecea, L. 1800

    Abstract

    The hypocretins (Hcrts), also known as orexins, are two neuropeptides produced exclusively in the lateral hypothalamus. They act on two specific receptors that are widely distributed across the brain and involved in a myriad of neurophysiological functions that include sleep, arousal, feeding, reward, fear, anxiety and cognition. Hcrt cell loss in humans leads to narcolepsy with cataplexy (narcolepsy type 1), a disorder characterized by intrusions of sleep into wakefulness, demonstrating that the Hcrt system is nonredundant and essential for sleep/wake stability. The causal link between Hcrts and arousal/wakefulness stabilisation has led to the development of a new class of drugs, Hcrt receptor antagonists to treat insomnia, based on the assumption that blocking orexin-induced arousal will facilitate sleep. This has been clinically validated: currently, two Hcrt receptor antagonists are approved to treat insomnia (suvorexant and lemborexant), with a New Drug Application recently submitted to the US Food and Drug Administration for a third drug (daridorexant). Other therapeutic applications under investigation include reduction of cravings in substance-use disorders and prevention of neurodegenerative disorders such as Alzheimer's disease, given the apparent bidirectional relationship between poor sleep and worsening of the disease. Circuit neuroscience findings suggest that the Hcrt system is a hub that integrates diverse inputs modulating arousal (e.g., circadian rhythms, metabolic status, positive and negative emotions) and conveys this information to multiple output regions. This neuronal architecture explains the wealth of physiological functions associated with Hcrts and highlights the potential of the Hcrt system as a therapeutic target for a number of disorders. We discuss present and future possible applications of drugs targeting the Hcrt system for the treatment of circuit-related neuropsychiatric and neurodegenerative conditions.

    View details for DOI 10.1111/joim.13406

    View details for PubMedID 35043499

  • Peripheral Lipopolyssacharide Rapidly Silences REM-Active LHGABA Neurons. Frontiers in behavioral neuroscience Borniger, J. C., de Lecea, L. 2021; 15: 649428

    Abstract

    Immune factors (e.g., cytokines, chemokines) can alter the activity of neuronal circuits to promote "sickness behavior," a suite of adaptive actions that organisms exhibit in response to infection/injury in order to maximize their chances of recovery (i.e., return to homeostasis). This includes drastic alterations in sleep/wake states, locomotor activity, and food intake, among other behaviors. Despite the ample evidence highlighting interactions between the brain and systemic immunity, studies on how immune challenges alter the activity of genetically defined cell populations controlling arousal states are scarce. As the lateral hypothalamus (LH) serves a major integrative function in behavioral arousal, food intake, and monitoring and responding to changes in systemic physiology, we investigated how GABAergic neurons within this brain region alter their activity across normal sleep/wake states and in response to a peripheral immune challenge with bacterial endotoxin [lipopolysaccharides (LPS)]. Using fiber photometry (GCaMP6s Ca2+ signal) in tandem with electroencephalogram (EEG)/EMG recordings to determine arousal states, we observed that population activity of GABAergic neurons in the lateral hypothalamus (LHGABA) is highest during rapid-eye-movement sleep (REM), and this activity changes drastically across spontaneous arousal state transitions, with the lowest activity observed during non-REM sleep. Upon intraperitoneal LPS challenge, LHGABA neurons rapidly decrease their activity in tandem with elimination of REM sleep behavior (characteristic of cytokine-induced sickness). Together, these data suggest that peripheral immune challenges can rapidly (in < 40 min) alter subcortical neuronal circuits controlling arousal states. Additionally, we demonstrate that fiber photometry offers a sensitive and cell-type specific tool that can be applied to study the neuronal substrates of sickness behavior.

    View details for DOI 10.3389/fnbeh.2021.649428

    View details for PubMedID 33716686

  • Twenty-Three Years of Hypocretins: The "Rosetta Stone" of Sleep/Arousal Circuits. Frontiers of neurology and neuroscience de Lecea, L. 2021; 45: 1-10

    Abstract

    The discovery of the hypocretins/orexins (HCRTs) has revolutionized sleep science in the last two decades. A combination of anatomical tracing methods, optogenetics, and pharmacology is delineating a blueprint of functional inputs and outputs of the HCRT system. Here, we discuss several models of HCRT action that involve the integration between physiological variables, circadian output, and sleep homeostasis. Generation of activity maps during the sleep-wake cycle at the cellular level will allow investigators to decipher computational frameworks modeling operations of HCRT networks.

    View details for DOI 10.1159/000514961

    View details for PubMedID 34052811

  • Heterogeneity of Hypocretin/Orexin Neurons. Frontiers of neurology and neuroscience Sagi, D., de Lecea, L., Appelbaum, L. 2021; 45: 61-74

    Abstract

    The multifunctional, hypothalamic hypocretin/orexin (HCRT)-producing neurons regulate an array of physiological and behavioral states including arousal, sleep, feeding, emotions, stress, and reward. How a presumably uniform HCRT neuron population regulates such a diverse set of functions is not clear. The role of the HCRT neuropeptides may vary depending on the timing and localization of secretion and neuronal activity. Moreover, HCRT neuropeptides may not mediate all functions ascribed to HCRT neurons. Some could be orchestrated by additional neurotransmitters and neuropeptides that are expressed in HCRT neurons. We hypothesize that HCRT neurons are segregated into genetically, anatomically and functionally distinct subpopulations. We discuss accumulating data that suggest the existence of such HCRT neuron subpopulations that may effectuate the diverse functions of these neurons in mammals and fish.

    View details for DOI 10.1159/000514964

    View details for PubMedID 34052814

  • Hypocretin/Orexins and Hyperarousal de Lecea, L. SPRINGERNATURE. 2020: 31
  • Neural and Hormonal Control of Sexual Behavior. Endocrinology Jennings, K. J., de Lecea, L. 2020

    Abstract

    Gonadal hormones contribute to the sexual differentiation of brain and behavior throughout the lifespan, from initial neural patterning to 'activation' of adult circuits. Sexual behavior is an ideal system in which to investigate the mechanisms underlying hormonal activation of neural circuits. Sexual behavior is a hormonally regulated, innate social behavior found across species. Although both sexes seek out and engage in sexual behavior, the specific actions involved in mating are sexually dimorphic. Thus, the neural circuits mediating sexual motivation and behavior in males and females are overlapping yet distinct. Furthermore, sexual behavior is strongly dependent on circulating gonadal hormones in both sexes. There has been significant recent progress on elucidating how gonadal hormones modulate physiological properties within sexual behavior circuits with consequences for behavior. Therefore, in this mini-review we review the neural circuits of male and female sexual motivation and behavior from initial sensory detection of pheromones to the extended amygdala and on to medial hypothalamic nuclei and reward systems. We also discuss how gonadal hormones impact the physiology and functioning of each node within these circuits. By better understanding the myriad of ways in which gonadal hormones impact sexual behavior circuits, we can gain a richer and more complete appreciation for the neural substrates of complex behavior.

    View details for DOI 10.1210/endocr/bqaa150

    View details for PubMedID 32845294

  • Impaired hypocretin/orexin system alters responses to salient stimuli in obese male mice. The Journal of clinical investigation Tan, Y., Hang, F., Liu, Z., Stoiljkovic, M., Wu, M., Tu, Y., Han, W., Lee, A. M., Kelley, C., Hajos, M., Lu, L., de Lecea, L., de Araujo, I., Picciotto, M., Horvath, T. L., Gao, X. 2020

    Abstract

    The brain has evolved in an environment where food sources are scarce and foraging for food is one of the major challenges for survival of the individual and species. Basic and clinical studies show that obesity/overnutrition leads to overwhelming changes in the brain in animals and humans. However, the exact mechanisms underlying the consequences of excessive energy intake are not well understood. Neurons expressing the neuropeptide hypocretin/orexin (Hcrt) in the lateral/perifonical hypothalamus (LH) are critical for homeostatic regulation, reward seeking, stress response, and cognitive functions. In this study, we examined adaptations in Hcrt cells regulating behavioral responses to salient stimuli in diet-induced obese mice. Our results demonstrated changes in primary cilia, synaptic transmission and plasticity, cellular responses to neurotransmitters necessary for reward seeking and stress responses in Hcrt neurons from obese mice. Activities of neuronal networks in the LH and hippocampus were impaired as a result of decreased hypocretinergic function. The weakened Hcrt system decreased reward seeking while altering responses to acute stress (stress coping strategy), which were reversed by selectively activating Hcrt cells with chemogenetics. Taken together, our data suggest that a deficiency in the Hcrt signaling may be a common cause of behavioral changes (such as lowered arousal, weakened reward seek and altered stress response) in obese animals.

    View details for DOI 10.1172/JCI130889

    View details for PubMedID 32516139

  • Editorial: Hypocretins/Orexins. Frontiers in endocrinology Lopez, M., de Lecea, L., Dieguez, C. 2020; 11: 357

    View details for DOI 10.3389/fendo.2020.00357

    View details for PubMedID 32582031

  • The hypocretin (orexin) system: from a neural circuitry perspective. Neuropharmacology Li, S. B., de Lecea, L. n. 2020; 167: 107993

    Abstract

    Hypocretin/orexin neurons are distributed restrictively in the hypothalamus, a brain region known to orchestrate diverse functions including sleep, reward processing, food intake, thermogenesis, and mood. Since the hypocretins/orexins were discovered more than two decades ago, extensive studies have accumulated concrete evidence showing the pivotal role of hypocretin/orexin in diverse neural modulation. New method of viral-mediated tracing system offers the possibility to map the monosynaptic inputs and detailed anatomical connectivity of Hcrt neurons. With the development of powerful research techniques including optogenetics, fiber-photometry, cell-type/pathway specific manipulation and neuronal activity monitoring, as well as single-cell RNA sequencing, the details of how hypocretinergic system execute functional modulation of various behaviors are coming to light. In this review, we focus on the function of neural pathways from hypocretin neurons to target brain regions. Anatomical and functional inputs to hypocretin neurons are also discussed. We further briefly summarize the development of pharmaceutical compounds targeting hypocretin signaling. This article is part of the special issue on Neuropeptides.

    View details for DOI 10.1016/j.neuropharm.2020.107993

    View details for PubMedID 32135427

  • Neurobiological and Hormonal Mechanisms Regulating Women's Sleep. Frontiers in neuroscience Dorsey, A., de Lecea, L., Jennings, K. J. 2020; 14: 625397

    Abstract

    Sleep is crucial for optimal well-being, and sex differences in sleep quality have significant implications for women's health. We review the current literature on sex differences in sleep, such as differences in objective and subjective sleep measures and their relationship with aging. We then discuss the convincing evidence for the role of ovarian hormones in regulating female sleep, and survey how these hormones act on a multitude of brain regions and neurochemicals to impact sleep. Lastly, we identify several important areas in need of future research to narrow the knowledge gap and improve the health of women and other understudied populations.

    View details for DOI 10.3389/fnins.2020.625397

    View details for PubMedID 33519372

  • Hypothalamic circuitry underlying stress-induced insomnia and peripheral immunosuppression Science Advances Li, S. B., Borniger, J. C., Yamaguchi, H., Hédou, J., Gaudilliere, B., de Lecea, L. 2020; 6 (37)

    View details for DOI 10.1126/sciadv.abc2590

  • Sleep and neuropsychiatric illness NEUROPSYCHOPHARMACOLOGY Winkelman, J. W., de Lecea, L. 2020; 45 (1): 1–2

    View details for DOI 10.1038/s41386-019-0514-5

    View details for Web of Science ID 000499141800001

    View details for PubMedID 31486776

    View details for PubMedCentralID PMC6879507

  • Brain Circuit of Claustrophobia-like Behavior in Mice Identified by Upstream Tracing of Sighing. Cell reports Li, P. n., Li, S. B., Wang, X. n., Phillips, C. D., Schwarz, L. A., Luo, L. n., de Lecea, L. n., Krasnow, M. A. 2020; 31 (11): 107779

    Abstract

    Emotions are distinct patterns of behavioral and physiological responses triggered by stimuli that induce different brain states. Elucidating the circuits is difficult because of challenges in interrogating emotional brain states and their complex outputs. Here, we leverage the recent discovery in mice of a neural circuit for sighing, a simple, quantifiable output of various emotions. We show that mouse confinement triggers sighing, and this "claustrophobic" sighing, but not accompanying tachypnea, requires the same medullary neuromedin B (Nmb)-expressing neurons as physiological sighing. Retrograde tracing from the Nmb neurons identified 12 forebrain centers providing presynaptic input, including hypocretin (Hcrt)-expressing lateral hypothalamic neurons. Confinement activates Hcrt neurons, and optogenetic activation induces sighing and tachypnea whereas pharmacologic inhibition suppresses both responses. The effect on sighing is mediated by HCRT directly on Nmbneurons. We propose that this HCRT-NMB neuropeptide relay circuit mediates claustrophobic sighing and that activated Hcrt neurons are a claustrophobia brain state that directly controls claustrophobic outputs.

    View details for DOI 10.1016/j.celrep.2020.107779

    View details for PubMedID 32553161

  • Arousal-state dependent alterations in VTA-GABAergic neuronal activity. eNeuro Eban-Rothschild, A. n., Borniger, J. C., Rothschild, G. n., Giardino, W. J., Morrow, J. G., de Lecea, L. n. 2020

    Abstract

    Decades of research have implicated the ventral tegmental area (VTA) in motivation, learning and reward processing. We and others recently demonstrated that it also serves as an important node in sleep/wake regulation. Specifically, VTA-dopaminergic neuron activation is sufficient to drive wakefulness and necessary for the maintenance of wakefulness. However, the role of VTA-GABAergic neurons in arousal regulation is not fully understood. It is still unclear whether VTA-GABAergic neurons predictably alter their activity across arousal states, what is the nature of interactions between VTA-GABAergic activity and cortical oscillations, and how activity in VTA-GABAergic neurons relates to VTA-dopaminergic neurons in the context of sleep/wake regulation. To address these, we simultaneously recorded population activity from VTA-subpopulations and EEG/EMG signals during spontaneous sleep/wake states and in the presence of salient stimuli in freely-behaving mice. We found that VTA-GABAergic neurons exhibit robust arousal-state-dependent alterations in population activity, with high activity and transients during wakefulness and REM sleep. During wakefulness, population activity of VTA-GABAergic neurons, but not VTA-dopaminergic neurons, was positively correlated with EEG gamma power and negatively correlated with theta power. During NREM sleep, population activity in both VTA-GABAergic and VTA-dopaminergic neurons negatively correlated with delta, theta, and sigma power bands. Salient stimuli, with both positive and negative valence, activated VTA-GABAergic neurons. Together, our data indicate that VTA-GABAergic neurons, like their dopaminergic counterparts, drastically alter their activity across sleep-wake states. Changes in their activity predicts cortical oscillatory patterns reflected in the EEG, which are distinct from EEG spectra associated with dopaminergic neural activity.Statement of Significance Little is known about how ventral tegmental area (VTA) neural ensembles couple arousal to motivated behaviors. Using cell-type specific genetic tools, we investigated the population activity of GABAergic and dopaminergic neurons within the VTA across sleep/wake states and in the presence of salient stimuli. We demonstrate that coordinated neural activity within VTA-GABAergic neurons peaks during wakefulness and REM sleep. Furthermore, neuronal activity in VTA-GABAergic neurons is correlated with high frequency, low amplitude cortical oscillations during waking, but negatively correlated with high amplitude slower frequency oscillations during NREM sleep. Our results demonstrate that VTA-GABAergic neuronal activity is tightly linked to cortical arousal and highlight this population as a potential important node in sleep/wake regulation.

    View details for DOI 10.1523/ENEURO.0356-19.2020

    View details for PubMedID 32054621

  • Multisensory modulation of body ownership in mice. Neuroscience of consciousness Buckmaster, C. L., Rathmann-Bloch, J. E., de Lecea, L. n., Schatzberg, A. F., Lyons, D. M. 2020; 2020 (1): niz019

    Abstract

    Body ownership is a fundamental aspect of self-consciousness that reflects more than the presence of physical body parts. As demonstrated by the rubber hand illusion (RHI), human brains construct body ownership experiences using available multisensory information. Experimental conditions similar to those that induce the RHI in humans have been recently adapted to induce the rubber tail illusion (RTI) in mice. Here, we show that the RTI is enhanced in both sexes of mice by repetitive synchronous stroking comprised of correlated visual and tactile stimulation of real and rubber tails compared to visual-only mimicked stroking conducted without tactile stimulation. The RTI also appears to be enhanced in female but not male mice by slow compared to fast stroking that reflects an interoceptive manipulation associated with affective touch in humans. Sex differences in slow stroking effects are exploratory and require replication in mice. Sex differences have not been reported for the RHI in healthy humans, but women rate slow stroking as more affectively pleasant compared to the ratings of men. Results suggest that the RHI in humans resembles aspects of the RTI in mice. Studies of mice may therefore provide neurobiological insights on evolutionarily conserved mechanisms of bodily self-consciousness in humans.

    View details for DOI 10.1093/nc/niz019

    View details for PubMedID 31988796

    View details for PubMedCentralID PMC6977007

  • CRISPR/Cas9 Editing of Neuropeptide Receptor Signaling Reveals an Extended Amygdala Circuit Mechanism Modulating Alcohol Drinking, Anxiety, and Avoidance Giardino, W., Yamaguchi, H., de Lecea, L. NATURE PUBLISHING GROUP. 2019: 505
  • Construction of Viral Vectors for Cell Type-specific CRISPR Gene Editing in the Adult Mouse Brain BIO-PROTOCOL Yamaguchi, H., de Lecea, L. 2019; 9 (16)
  • Construction of Viral Vectors for Cell Type-specific CRISPR Gene Editing in the Adult Mouse Brain. Bio-protocol Yamaguchi, H., de Lecea, L. 2019; 9 (16): e3334

    Abstract

    Recently developed gene editing technologies based on engineered CRISPR/Cas9 systems enables researchers to disrupt genes in a cell type-specific manner in the adult mouse brain. Using these technologies, we recently showed that the dopamine beta-hydroxylase gene in Locus Coeruleus (LC) norepinephrine neurons plays a vital role in the maintenance of wakefulness. Our method consists of four steps, (1) crossing Cre-dependent spCas9 knockin mice with a Cre-driver mouse line to express spCas9 in the target neural populations, (2) cloning of sgRNA, (3) construction of an AAV (adeno associated virus) vector expressing dual sgRNA, and (4) virus packaging and stereotaxic injection of the virus into the target brain area. Here, we describe a detailed protocol of AAV vector construction for cell type-specific CRISPR gene editing in the adult mouse brain. The method adopts a dual-sgRNA strategy for efficient disruption of the target gene. At first, a few different sgRNAs targeting the same gene are cloned into a plasmid expressing spCas9. After evaluation of the sgRNAs by a T7 endonuclease assay, the two most efficient sgRNAs are cloned in tandem into an AAV vector using the Gibson Assembly method.

    View details for DOI 10.21769/BioProtoc.3334

    View details for PubMedID 33654839

    View details for PubMedCentralID PMC7854134

  • In vivo cell type-specific CRISPR gene editing for sleep research JOURNAL OF NEUROSCIENCE METHODS Yamaguchi, H., de Lecea, L. 2019; 316: 99–102
  • Hypocretin and the Regulation of Sleep-Wake Transitions HANDBOOK OF SLEEP RESEARCH, VOL 30 Nevarez, N., de Lecea, L., Dringenberg, H. C. 2019; 30: 89–99
  • In vivo cell type-specific CRISPR knockdown of dopamine beta hydroxylase reduces locus coeruleus evoked wakefulness. Nature communications Yamaguchi, H., Hopf, F. W., Li, S. B., de Lecea, L. 2018; 9 (1): 5211

    Abstract

    Locus coeruleus (LC) neurons in the brainstem have long been associated with attention and arousal. Optogenetic stimulation of LC-NE neurons induces immediate sleep-to-wake transitions. However, LC neurons also secrete other neurotransmitters in addition to NE. To interrogate the role of NE derived from the LC in regulating wakefulness, we applied in vivo cell type-specific CRISPR/Cas9 technology to disrupt the dopamine beta hydroxylase (dbh) gene selectively in adult LC-NE neurons. Unilateral dbh gene disruption abolished immediate arousal following optogenetic stimulation of LC. Bilateral LC-specific dbh disruption significantly reduced NE concentration in LC projection areas and reduced wake length even in the presence of salient stimuli. These results suggest that NE may be crucial for the awakening effect of LC stimulation and serve as proof-of-principle that CRISPR gene editing in adult neurons can be used to interrogate gene function within genetically-defined neuronal circuitry associated with complex behaviors.

    View details for DOI 10.1038/s41467-018-07566-3

    View details for PubMedID 30523254

    View details for PubMedCentralID PMC6283864

  • Parallel Circuits From the Bed Nuclei of Stria Terminalis to the Lateral Hypothalamus Drive Opposing Emotional States Giardino, W., Eban-Rothschild, A., Christoffel, D., Li, S., Malenka, R., de Lecea, L. NATURE PUBLISHING GROUP. 2018: S234
  • In vivo cell type-specific CRISPR gene editing for sleep research. Journal of neuroscience methods Yamaguchi, H., de Lecea, L. 2018

    Abstract

    Sleep is an innate behavior conserved in all animals and, in vertebrates, is regulated by neuronal circuits in the brain. The conventional techniques of forward and reverse genetics have enabled researchers to investigate the molecular mechanisms that regulate sleep and arousal. However, functional interrogation of genes in specific cell subtypes in the brain remains a challenge. Here, we review the background of newly developed gene-editing technologies using engineered CRISPR/Cas9 system and describe the application to interrogate gene functions within genetically-defined brain cell populations in sleep research.

    View details for PubMedID 30439390

  • Parallel circuits from the bed nuclei of stria terminalis to the lateral hypothalamus drive opposing emotional states. Nature neuroscience Giardino, W. J., Eban-Rothschild, A., Christoffel, D. J., Li, S., Malenka, R. C., de Lecea, L. 2018

    Abstract

    Lateral hypothalamus (LH) neurons containing the neuropeptide hypocretin (HCRT; orexin) modulate affective components of arousal, but their relevant synaptic inputs remain poorly defined. Here we identified inputs onto LH neurons that originate from neuronal populations in the bed nuclei of stria terminalis (BNST; a heterogeneous region of extended amygdala). We characterized two non-overlapping LH-projecting GABAergic BNST subpopulations that express distinct neuropeptides (corticotropin-releasing factor, CRF, and cholecystokinin, CCK). To functionally interrogate BNSTLH circuitry, we used tools for monitoring and manipulating neural activity with cell-type-specific resolution in freely behaving mice. We found that Crf-BNST and Cck-BNST neurons respectively provide abundant and sparse inputs onto Hcrt-LH neurons, display discrete physiological responses to salient stimuli, drive opposite emotionally valenced behaviors, and receive different proportions of inputs from upstream networks. Together, our data provide an advanced model for how parallel BNSTLH pathways promote divergent emotional states via connectivity patterns of genetically defined, circuit-specific neuronal subpopulations.

    View details for PubMedID 30038273

  • Hypocretin as a Hub for Arousal and Motivation FRONTIERS IN NEUROLOGY Tyree, S. M., Bomiger, J. C., de Lecea, L. 2018; 9: 413

    Abstract

    The lateral hypothalamus is comprised of a heterogeneous mix of neurons that serve to integrate and regulate sleep, feeding, stress, energy balance, reward, and motivated behavior. Within these populations, the hypocretin/orexin neurons are among the most well studied. Here, we provide an overview on how these neurons act as a central hub integrating sensory and physiological information to tune arousal and motivated behavior accordingly. We give special attention to their role in sleep-wake states and conditions of hyper-arousal, as is the case with stress-induced anxiety. We further discuss their roles in feeding, drug-seeking, and sexual behavior, which are all dependent on the motivational state of the animal. We further emphasize the application of powerful techniques, such as optogenetics, chemogenetics, and fiber photometry, to delineate the role these neurons play in lateral hypothalamic functions.

    View details for PubMedID 29928253

  • Hypocretin/orexin deficiency decreases cocaine abuse liability NEUROPHARMACOLOGY Steiner, N., Rossetti, C., Sakurai, T., Yanagisawa, M., de Lecea, L., Magistretti, P. J., Halfon, O., Boutrel, B. 2018; 133: 395–403

    Abstract

    Compelling evidence indicates that hypocretin/orexin signaling regulates arousal, stress and reward-seeking behaviors. However, most studies on drug reward-related processes have so far described the effects of pharmacological blockers disrupting hypocretin/orexin transmission. We report here an extensive study on cocaine-related behaviors in hypocretin/orexin-deficient mice (KO) and their heterozygous (HET) and wildtype (WT) littermates. We evaluated behavioral sensitization following repeated administrations and preference for an environment repeatedly paired with cocaine injections (15 mg/kg). Mice were also trained to self-administer cocaine (0.5-1.5 mg/kg/infusion). Our observations show that whereas all mice exhibited quite similar responses to acute administration of cocaine, only Hcrt KO mice exhibited reduced cocaine-seeking behaviors following a period of abstinence or extinction, and reduced cocaine incubation craving. Further, if the present findings confirm that Hcrt deficient mice may display a hypoactive phenotype, possibly linked to a reduced alertness concomitant to a decreased exploration of their environment, hypocretin/orexin defiency did not cause any attentional deficit. We thus report that innate disruption of hypocretin/orexin signaling moderately alters cocaine reward but significantly reduces long-term affective dependence that may explain the lack of relapse for cocaine seeking seen in Hcrt KO mice. Overall, with blunted cocaine intake at the highest concentration and reduced responsiveness to cocaine cues after prolonged abstinence, our findings suggest that hypocretin deficient mice may display signs of resilience to cocaine addiction.

    View details for PubMedID 29454841

  • Neuronal Mechanisms for Sleep/Wake Regulation and Modulatory Drive NEUROPSYCHOPHARMACOLOGY Eban-Rothschild, A., Appelbaum, L., de Lecea, L. 2018; 43 (5): 937–52

    Abstract

    Humans have been fascinated by sleep for millennia. After almost a century of scientific interrogation, significant progress has been made in understanding the neuronal regulation and functions of sleep. The application of new methods in neuroscience that enable the analysis of genetically defined neuronal circuits with unprecedented specificity and precision has been paramount in this endeavor. In this review, we first discuss electrophysiological and behavioral features of sleep/wake states and the principal neuronal populations involved in their regulation. Next, we describe the main modulatory drives of sleep and wakefulness, including homeostatic, circadian, and motivational processes. Finally, we describe a revised integrative model for sleep/wake regulation.

    View details for PubMedID 29206811

    View details for PubMedCentralID PMC5854814

  • Optical Probing of Orexin/Hypocretin Receptor Antagonists. Sleep Li, S. B., Nevárez, N. n., Giardino, W. J., de Lecea, L. n. 2018

    Abstract

    The present study investigated the function of Hypocretin (Hcrt or Orexin/OX) receptor antagonists in sleep modulation and memory function with optical methods in transgenic mice.We used Hcrt-IRES-Cre knock-in mice and AAV vectors expressing channelrhodopsin-2 (ChR2) to render Hcrt neurons sensitive to blue light stimulation. We optogenetically stimulated Hcrt neurons and measured latencies to wakefulness in the presence or absence of OX1/2R antagonists and Zolpidem. We also examined endogenous Hcrt neuronal activity with fiber photometry. Changes in memory after optogenetic sleep disruption were evaluated by the novel object recognition test (NOR) and compared for groups treated with vehicle, OX1/2R antagonists, or Zolpidem. We also analyzed EEG power spectra of wakefulness, rapid eye movement (REM) sleep, and non-REM (NREM) sleep following the injections of vehicle, OX1/2R antagonists, and Zolpidem in young adult mice.Acute optogenetic stimulation of Hcrt neurons at different frequencies resulted in wakefulness. Treatment with dual OX1/2R antagonists (DORAs) DORA12 and MK6096, as well as selective OX2R antagonist MK1064 and Zolpidem, but not selective OX1R antagonist 1SORA1, significantly reduced the bout length of optogenetic stimulation-evoked wakefulness episode. Fiber photometry recordings of GCaMP6f signals showed that Hcrt neurons are active during wakefulness, even in the presence of OXR antagonists. Treatment with dual OX1/2R antagonists improved memory function despite optogenetic sleep fragmentation caused impaired memory function in a NOR test.Our results show DORAs and selective OX2R antagonists stabilize sleep and improve sleep-dependent cognitive processes even when challenged by optogenetic stimulation mimicking highly arousing stimuli.

    View details for PubMedID 30060151

  • Optical Probing of Orexin/Hypocretin Receptor Antagonists Sleep Li, S., Nevárez, N., Giardino, W. J., de Lecea, L. 2018

    View details for DOI 10.1093/sleep/zsy141

  • In vivo cell type-specific CRISPR knockdown of dopamine beta hydroxylase reduces locus coeruleus evoked wakefulness Nature Communications Yamaguchi, H., Hopf, F., Li, S., de Lecea, L. 2018; 9
  • Recent advances in understanding the roles of hypocretin/orexin in arousal, affect, and motivation. F1000Research Nevarez, N., de Lecea, L. 2018; 7

    Abstract

    The hypocretins (Hcrts) are two alternatively spliced neuropeptides (Hcrt1/Ox-A and Hcrt2/Ox-B) that are synthesized exclusively in the hypothalamus. Data collected in the 20 years since their discovery have supported the view that the Hcrts play a broad role in the control of arousal with a particularly important role in the maintenance of wakefulness and sleep-to-wake transitions. While this latter point has received an overwhelming amount of research attention, a growing literature has begun to broaden our understanding of the many diverse roles that the Hcrts play in physiology and behavior. Here, we review recent advances in the neurobiology of Hcrt in three sections. We begin by surveying findings on Hcrt function within normal sleep/wake states as well as situations of aberrant sleep (that is, narcolepsy). In the second section, we discuss research establishing a role for Hcrt in mood and affect (that is, anxiety, stress, and motivation). Finally, in the third section, we briefly discuss future directions for the field and place an emphasis on analytical modeling of Hcrt neural activity. We hope that the data discussed here provide a broad overview of recent progress in the field and make clear the diversity of roles played by these neuromodulators.

    View details for PubMedID 30254737

  • Hypothalamic Tuberomammillary Nucleus Neurons: Electrophysiological Diversity and Essential Role in Arousal Stability JOURNAL OF NEUROSCIENCE Fujita, A., Bonnavion, P., Wilson, M. H., Mickelsen, L. E., Bloit, J., de Lecea, L., Jackson, A. C. 2017; 37 (39): 9574–92

    Abstract

    Histaminergic (HA) neurons, found in the posterior hypothalamic tuberomammillary nucleus (TMN), extend fibers throughout the brain and exert modulatory influence over numerous physiological systems. Multiple lines of evidence suggest that the activity of HA neurons is important in the regulation of vigilance despite the lack of direct, causal evidence demonstrating its requirement for the maintenance of arousal during wakefulness. Given the strong correlation between HA neuron excitability and behavioral arousal, we investigated both the electrophysiological diversity of HA neurons in brain slices and the effect of their acute silencing in vivo in male mice. For this purpose, we first validated a transgenic mouse line expressing cre recombinase in histidine decarboxylase-expressing neurons (Hdc-Cre) followed by a systematic census of the membrane properties of both HA and non-HA neurons in the ventral TMN (TMNv) region. Through unsupervised hierarchical cluster analysis, we found electrophysiological diversity both between TMNv HA and non-HA neurons, and among HA neurons. To directly determine the impact of acute cessation of HA neuron activity on sleep-wake states in awake and behaving mice, we examined the effects of optogenetic silencing of TMNv HA neurons in vivo We found that acute silencing of HA neurons during wakefulness promotes slow-wave sleep, but not rapid eye movement sleep, during a period of low sleep pressure. Together, these data suggest that the tonic firing of HA neurons is necessary for the maintenance of wakefulness, and their silencing not only impairs arousal but is sufficient to rapidly and selectively induce slow-wave sleep.SIGNIFICANCE STATEMENT The function of monoaminergic systems and circuits that regulate sleep and wakefulness is often disrupted as part of the pathophysiology of many neuropsychiatric disorders. One such circuit is the posterior hypothalamic histamine (HA) system, implicated in supporting wakefulness and higher brain function, but has been difficult to selectively manipulate owing to cellular heterogeneity in this region. Here we use a transgenic mouse to interrogate both the characteristic firing properties of HA neurons and their specific role in maintaining wakefulness. Our results demonstrate that the acute, cell type-specific silencing of HA neurons during wakefulness is sufficient to not only impair arousal but to rapidly and selectively induce slow-wave sleep. This work furthers our understanding of HA-mediated mechanisms that regulate behavioral arousal.

    View details for PubMedID 28874450

  • To sleep or not to sleep: neuronal and ecological insights. Current opinion in neurobiology Eban-Rothschild, A., Giardino, W. J., de Lecea, L. 2017; 44: 132-138

    Abstract

    Daily, animals need to decide when to stop engaging in cognitive processes and behavioral responses to the environment, and go to sleep. The main processes regulating the daily organization of sleep and wakefulness are circadian rhythms and homeostatic sleep pressure. In addition, motivational processes such as food seeking and predator evasion can modulate sleep/wake behaviors. Here, we discuss the principal processes regulating the propensity to stay awake or go to sleep-focusing on neuronal and behavioral aspects. We first introduce the neuronal populations involved in sleep/wake regulation. Next, we describe the circadian and homeostatic drives for sleep. Then, we highlight studies demonstrating various effects of motivational processes on sleep/wake behaviors, and discuss possible neuronal mechanisms underlying their control.

    View details for DOI 10.1016/j.conb.2017.04.010

    View details for PubMedID 28500869

  • Neuronal substrates for initiation, maintenance, and structural organization of sleep/wake states. F1000Research Eban-Rothschild, A., de Lecea, L. 2017; 6: 212-?

    Abstract

    Animals continuously alternate between sleep and wake states throughout their life. The daily organization of sleep and wakefulness is orchestrated by circadian, homeostatic, and motivational processes. Over the last decades, much progress has been made toward determining the neuronal populations involved in sleep/wake regulation. Here, we will discuss how the application of advanced in vivo tools for cell type-specific manipulations now permits the functional interrogation of different features of sleep/wake state regulation: initiation, maintenance, and structural organization. We will specifically focus on recent studies examining the roles of wake-promoting neuronal populations.

    View details for DOI 10.12688/f1000research.9677.1

    View details for PubMedID 28357049

  • Stress Coping and Resilience Modeled in Mice ANIMAL MODELS FOR THE STUDY OF HUMAN DISEASE, 2ND EDITION Lyons, D. M., de Lecea, L., Schatzberg, A. F., Conn, P. M. 2017: 1145-1153
  • Behavioral Neuroscience of Orexin/Hypocretin Preface BEHAVIORAL NEUROSCIENCE OF OREXIN/HYPOCRETIN Lawrence, A. J., de Lecea, L., Lawrence, A. J., DeLecea, L. 2017; 33: V-VI
  • Lateral Hypothalamic Control of the Ventral Tegmental Area: Reward Evaluation and the Driving of Motivated Behavior. Frontiers in systems neuroscience Tyree, S. M., de Lecea, L. n. 2017; 11: 50

    Abstract

    The lateral hypothalamus (LH) plays an important role in many motivated behaviors, sleep-wake states, food intake, drug-seeking, energy balance, etc. It is also home to a heterogeneous population of neurons that express and co-express multiple neuropeptides including hypocretin (Hcrt), melanin-concentrating hormone (MCH), cocaine- and amphetamine-regulated transcript (CART) and neurotensin (NT). These neurons project widely throughout the brain to areas such as the locus coeruleus, the bed nucleus of the stria terminalis, the amygdala and the ventral tegmental area (VTA). Lateral hypothalamic projections to the VTA are believed to be important for driving behavior due to the involvement of dopaminergic reward circuitry. The purpose of this article is to review current knowledge regarding the lateral hypothalamic connections to the VTA and the role they play in driving these behaviors.

    View details for DOI 10.3389/fnsys.2017.00050

    View details for PubMedID 28729827

    View details for PubMedCentralID PMC5498520

  • Rat intersubjective decisions are encoded by frequency-specific oscillatory contexts. Brain and behavior Schaich Borg, J. n., Srivastava, S. n., Lin, L. n., Heffner, J. n., Dunson, D. n., Dzirasa, K. n., de Lecea, L. n. 2017; 7 (6): e00710

    Abstract

    It is unknown how the brain coordinates decisions to withstand personal costs in order to prevent other individuals' distress. Here we test whether local field potential (LFP) oscillations between brain regions create "neural contexts" that select specific brain functions and encode the outcomes of these types of intersubjective decisions.Rats participated in an "Intersubjective Avoidance Test" (IAT) that tested rats' willingness to enter an innately aversive chamber to prevent another rat from getting shocked. c-Fos immunoreactivity was used to screen for brain regions involved in IAT performance. Multi-site local field potential (LFP) recordings were collected simultaneously and bilaterally from five brain regions implicated in the c-Fos studies while rats made decisions in the IAT. Local field potential recordings were analyzed using an elastic net penalized regression framework.Rats voluntarily entered an innately aversive chamber to prevent another rat from getting shocked, and c-Fos immunoreactivity in brain regions known to be involved in human empathy-including the anterior cingulate, insula, orbital frontal cortex, and amygdala-correlated with the magnitude of "intersubjective avoidance" each rat displayed. Local field potential recordings revealed that optimal accounts of rats' performance in the task require specific frequencies of LFP oscillations between brain regions in addition to specific frequencies of LFP oscillations within brain regions. Alpha and low gamma coherence between spatially distributed brain regions predicts more intersubjective avoidance, while theta and high gamma coherence between a separate subset of brain regions predicts less intersubjective avoidance. Phase relationship analyses indicated that choice-relevant coherence in the alpha range reflects information passed from the amygdala to cortical structures, while coherence in the theta range reflects information passed in the reverse direction.These results indicate that the frequency-specific "neural context" surrounding brain regions involved in social cognition encodes outcomes of decisions that affect others, above and beyond signals from any set of brain regions in isolation.

    View details for PubMedID 28638715

    View details for PubMedCentralID PMC5474713

  • Optogenetic Investigation of Arousal Circuits. International journal of molecular sciences Tyree, S. M., de Lecea, L. n. 2017; 18 (8)

    Abstract

    Modulation between sleep and wake states is controlled by a number of heterogeneous neuron populations. Due to the topological proximity and genetic co-localization of the neurons underlying sleep-wake state modulation optogenetic methods offer a significant improvement in the ability to benefit from both the precision of genetic targeting and millisecond temporal control. Beginning with an overview of the neuron populations mediating arousal, this review outlines the progress that has been made in the investigation of arousal circuits since the incorporation of optogenetic techniques and the first in vivo application of optogenetic stimulation in hypocretin neurons in the lateral hypothalamus. This overview is followed by a discussion of the future progress that can be made by incorporating more recent technological developments into the research of neural circuits.

    View details for PubMedID 28809797

  • Hypocretins and Arousal. Current topics in behavioral neurosciences Li, S., Giardino, W. J., de Lecea, L. 2016

    Abstract

    How the brain controls vigilance state transitions remains to be fully understood. The discovery of hypocretins, also known as orexins, and their link to narcolepsy has undoubtedly allowed us to advance our knowledge on key mechanisms controlling the boundaries and transitions between sleep and wakefulness. Lack of function of hypocretin neurons (a relatively simple and non-redundant neuronal system) results in inappropriate control of sleep states without affecting the total amount of sleep or homeostatic mechanisms. Anatomical and functional evidence shows that the hypothalamic neurons that produce hypocretins/orexins project widely throughout the entire brain and interact with major neuromodulator systems in order to regulate physiological processes underlying wakefulness, attention, and emotions. Here, we review the role of hypocretins/orexins in arousal state transitions, and discuss possible mechanisms by which such a relatively small population of neurons controls fundamental brain state dynamics.

    View details for DOI 10.1007/7854_2016_58

    View details for PubMedID 28012091

  • VTA Dopaminergic Neurons Regulate Ethologically Relevant Sleep-Wake Behaviors Eban-Rothschild, A., Rothschild, G., Giardino, W. J., Jones, J. R., de Lecea, L. NATURE PUBLISHING GROUP. 2016: S259
  • Obesity- and gender-dependent role of endogenous somatostatin and cortistatin in the regulation of endocrine and metabolic homeostasis in mice SCIENTIFIC REPORTS Luque, R. M., Cordoba-Chacon, J., Pozo-Salas, A. I., Porteiro, B., de Lecea, L., Nogueiras, R., Gahete, M. D., Castano, J. P. 2016; 6

    Abstract

    Somatostatin (SST) and cortistatin (CORT) regulate numerous endocrine secretions and their absence [knockout (KO)-models] causes important endocrine-metabolic alterations, including pituitary dysregulations. We have demonstrated that the metabolic phenotype of single or combined SST/CORT KO-models is not drastically altered under normal conditions. However, the biological actions of SST/CORT are conditioned by the metabolic-status (e.g. obesity). Therefore, we used male/female SST- and CORT-KO mice fed low-fat (LF) or high-fat (HF) diet to explore the interplay between SST/CORT and obesity in the control of relevant pituitary-axes and whole-body metabolism. Our results showed that the SST/CORT role in the control of GH/prolactin secretions is maintained under LF- and HF-diet conditions as SST-KOs presented higher GH/prolactin-levels, while CORT-KOs displayed higher GH- and lower prolactin-levels than controls under both diets. Moreover, the impact of lack of SST/CORT on the metabolic-function was gender- and diet-dependent. Particularly, SST-KOs were more sensitive to HF-diet, exhibiting altered growth and body-composition (fat/lean percentage) and impaired glucose/insulin-metabolism, especially in males. Conversely, only males CORT-KO under LF-diet conditions exhibited significant alterations, displaying higher glucose-levels and insulin-resistance. Altogether, these data demonstrate a tight interplay between SST/CORT-axis and the metabolic status in the control of endocrine/metabolic functions and unveil a clear dissociation of SST/CORT roles.

    View details for DOI 10.1038/srep37992

    View details for Web of Science ID 000389199600001

    View details for PubMedID 27901064

    View details for PubMedCentralID PMC5128804

  • Hubs and spokes of the lateral hypothalamus: cell types, circuits and behaviour JOURNAL OF PHYSIOLOGY-LONDON Bonnavion, P., Mickelsen, L. E., Fujita, A., de Lecea, L., Jackson, A. C. 2016; 594 (22): 6443-6462

    Abstract

    The hypothalamus is among the most phylogenetically conserved regions in the vertebrate brain, reflecting its critical role in maintaining physiological and behavioural homeostasis. By integrating signals arising from both the brain and periphery, it governs a litany of behaviourally important functions essential for survival. In particular, the lateral hypothalamic area (LHA) is central to the orchestration of sleep-wake states, feeding, energy balance and motivated behaviour. Underlying these diverse functions is a heterogeneous assembly of cell populations typically defined by neurochemical markers, such as the well-described neuropeptides hypocretin/orexin and melanin-concentrating hormone. However, anatomical and functional evidence suggests a rich diversity of other cell populations with complex neurochemical profiles that include neuropeptides, receptors and components of fast neurotransmission. Collectively, the LHA acts as a hub for the integration of diverse central and peripheral signals and, through complex local and long-range output circuits, coordinates adaptive behavioural responses to the environment. Despite tremendous progress in our understanding of the LHA, defining the identity of functionally discrete LHA cell types, and their roles in driving complex behaviour, remain significant challenges in the field. In this review, we discuss advances in our understanding of the neurochemical and cellular heterogeneity of LHA neurons and the recent application of powerful new techniques, such as opto- and chemogenetics, in defining the role of LHA circuits in feeding, reward, arousal and stress. From pioneering work to recent developments, we review how the interrogation of LHA cells and circuits is contributing to a mechanistic understanding of how the LHA coordinates complex behaviour.

    View details for DOI 10.1113/JP271946

    View details for Web of Science ID 000389029900003

    View details for PubMedID 27302606

    View details for PubMedCentralID PMC5108896

  • Fasting modulates GH/IGF-I axis and its regulatory systems in the mammary gland of female mice: Influence of endogenous cortistatin. Molecular and cellular endocrinology Villa-Osaba, A., Gahete, M. D., Cordoba-Chacon, J., de Lecea, L., Castaño, J. P., Luque, R. M. 2016; 434: 14-24

    Abstract

    Growth hormone (GH) and insulin-like growth factor-I (IGF-I) are essential factors in mammary-gland (MG) development and are altered during fasting. However, no studies have investigated the alterations in the expression of GH/IGF-I and its regulatory systems (somatostatin/cortistatin and ghrelin) in MG during fasting. Therefore, this study was aimed at characterizing the regulation of GH/IGF-I/somatostatin/cortistatin/ghrelin-systems expression in MG of fasted female-mice (compared to fed-controls) and the influence of endogenous-cortistatin (using cortistatin-knockouts). Fasting decreased IGF-I while increased IGF-I/Insulin-receptors expression in MGs. Fasting provoked an increase in GH expression that might be associated to enhanced ghrelin-variants/ghrelin-O-acyl-transferase enzyme expression, while an upregulation of somatostatin-receptors was observed. However, cortistatin-knockouts mice showed a decrease in GH and somatostatin receptor-subtypes expression. Altogether, we demonstrate that GH/IGF-I, somatostatin/cortistatin and ghrelin systems expression is altered in MG during fasting, suggesting a relevant role in coordinating its response to metabolic stress, wherein endogenous cortistatin might be essential for an appropriate response.

    View details for DOI 10.1016/j.mce.2016.06.014

    View details for PubMedID 27291340

  • In vivo assessment of behavioral recovery and circulatory exchange in the peritoneal parabiosis model SCIENTIFIC REPORTS Castellano, J. M., Palner, M., Li, S., Freeman, G. M., Andy Nguyen, A., Shen, B., Stan, T., Mosher, K. I., Chin, F. T., de Lecea, L., Luo, J., Wyss-Coray, T. 2016; 6

    Abstract

    The sharing of circulation between two animals using a surgical procedure known as parabiosis has created a wealth of information towards our understanding of physiology, most recently in the neuroscience arena. The systemic milieu is a complex reservoir of tissues, immune cells, and circulating molecules that is surprisingly not well understood in terms of its communication across organ systems. While the model has been used to probe complex physiological questions for many years, critical parameters of recovery and exchange kinetics remain incompletely characterized, limiting the ability to design experiments and interpret results for complex questions. Here we provide evidence that mice joined by parabiosis gradually recover much physiology relevant to the study of brain function. Specifically, we describe the timecourse for a variety of recovery parameters, including those for general health and metabolism, motor coordination, activity, and sleep behavior. Finally, we describe the kinetics of chimerism for several lymphocyte populations as well as the uptake of small molecules into the brains of mice following parabiosis. Our characterization provides an important resource to those attempting to understand the complex interplay between the immune system and the brain as well as other organ systems.

    View details for DOI 10.1038/srep29015

    View details for Web of Science ID 000378851500002

    View details for PubMedID 27364522

    View details for PubMedCentralID PMC4929497

  • Cortistatin Is a Key Factor Regulating the Sex-Dependent Response of the GH and Stress Axes to Fasting in Mice ENDOCRINOLOGY Cordoba-Chacon, J., Gahete, M. D., Pozo-Salas, A. I., de Lecea, L., Castano, J. P., Luque, R. M. 2016; 157 (7): 2810-2823

    Abstract

    Cortistatin (CORT) shares high structural and functional similarities with somatostatin (SST) but displays unique sex-dependent pituitary actions. Indeed, although female CORT-knockout (CORT-KO) mice exhibit enhanced GH expression/secretion, Proopiomelanocortin expression, and circulating ACTH/corticosterone/ghrelin levels, male CORT-KO mice only display increased plasma GH/corticosterone levels. Changes in peripheral ghrelin and SST (rather than hypothalamic levels) seem to regulate GH/ACTH axes in CORT-KOs under fed conditions. Because changes in GH/ACTH axes during fasting provide important adaptive mechanisms, we sought to determine whether CORT absence influences GH/ACTH axes during fasting. Accordingly, fed and fasted male/female CORT-KO were compared with littermate controls. Fasting increased circulating GH levels in male/female controls but not in CORT-KO, suggesting that CORT can be a relevant regulator of GH secretion during fasting. However, GH levels were already higher in CORT-KO than in controls in fed state, which might preclude a further elevation in GH levels. Interestingly, although fasting-induced pituitary GH expression was elevated in both male/female controls, GH expression only increased in fasted female CORT-KOs, likely owing to specific changes observed in key factors controlling somatotrope responsiveness (ie, circulating ghrelin and IGF-1, and pituitary GHRH and ghrelin receptor expression). Fasting increased corticosterone levels in control and, most prominently, in CORT-KO mice, which might be associated with a desensitization to SST signaling and to an augmentation in CRH and ghrelin-signaling regulating corticotrope function. Altogether, these results provide compelling evidence that CORT plays a key, sex-dependent role in the regulation of the GH/ACTH axes in response to fasting.

    View details for DOI 10.1210/en.2016-1195

    View details for Web of Science ID 000378877200022

    View details for PubMedID 27175972

  • Hypocretin (Orexin) System and its Functions in Arousal/sleep de Lecea, L. ELSEVIER SCIENCE INC. 2016: 170S-171S
  • Superficial Layer-Specific Histaminergic Modulation of Medial Entorhinal Cortex Required for Spatial Learning CEREBRAL CORTEX He, C., Luo, F., Chen, X., Chen, F., Li, C., Ren, S., Qiao, Q., Zhang, J., de Lecea, L., Gao, D., Hu, Z. 2016; 26 (4): 1590-1608

    Abstract

    The medial entorhinal cortex (MEC) plays a crucial role in spatial learning and memory. Whereas the MEC receives a dense histaminergic innervation from the tuberomamillary nucleus of the hypothalamus, the functions of histamine in this brain region remain unclear. Here, we show that histamine acts via H1Rs to directly depolarize the principal neurons in the superficial, but not deep, layers of the MEC when recording at somata. Moreover, histamine decreases the spontaneous GABA, but not glutamate, release onto principal neurons in the superficial layers by acting at presynaptic H3Rs without effect on synaptic release in the deep layers. Histamine-induced depolarization is mediated via inhibition of Kir channels and requires the activation of protein kinase C, whereas the inhibition of spontaneous GABA release by histamine depends on voltage-gated Ca(2+) channels and extracellular Ca(2+). Furthermore, microinjection of the H1R or H3R, but not H2R, antagonist respectively into the superficial, but not deep, layers of MEC impairs rat spatial learning as assessed by water maze tasks but does not affect the motor function and exploratory activity in an open field. Together, our study indicates that histamine plays an essential role in spatial learning by selectively regulating neuronal excitability and synaptic transmission in the superficial layers of the MEC.

    View details for DOI 10.1093/cercor/bhu322

    View details for Web of Science ID 000374246700021

    View details for PubMedID 25595181

  • Resting easy with a sleep regulator ELIFE Giardino, W. J., de Lecea, L. 2015; 4

    View details for DOI 10.7554/eLife.12093

    View details for Web of Science ID 000367511500001

    View details for PubMedID 26651823

    View details for PubMedCentralID PMC4744186

  • Sleep disruption impairs haematopoietic stem cell transplantation in mice NATURE COMMUNICATIONS Rolls, A., Pang, W. W., Ibarra, I., Colas, D., Bonnavion, P., Korin, B., Heller, H. C., Weissman, I. L., de Lecea, L. 2015; 6

    Abstract

    Many of the factors affecting the success of haematopoietic cell transplantation are still unknown. Here we show in mice that donor sleep deprivation reduces the ability of its haematopoietic stem cells (HSCs) to engraft and reconstitute the blood and bone marrow of an irradiated recipient by more than 50%. We demonstrate that sleep deprivation downregulates the expression of microRNA (miR)-19b, a negative regulator of the suppressor of cytokine signalling (SOCS) genes, which inhibit HSC migration and homing. Accordingly, HSCs from sleep-deprived mice have higher levels of SOCS genes expression, lower migration capacity in vitro and reduced homing to the bone marrow in vivo. Recovery of sleep after sleep deprivation restored the reconstitution potential of the HSCs. Taken together, this study provides insights into cellular and molecular mechanisms underlying the effects of sleep deprivation on HSCs, emphasizing the potentially critical role of donor sleep in the success of bone marrow transplantation.

    View details for DOI 10.1038/ncomms9516

    View details for Web of Science ID 000364930800001

    View details for PubMedID 26465715

    View details for PubMedCentralID PMC4621781

  • Not So Giants: Mice Lacking Both Somatostatin and Cortistatin Have High GH Levels but Show No Changes in Growth Rate or IGF-1 Levels ENDOCRINOLOGY Pedraza-Arevalo, S., Cordoba-Chacon, J., Pozo-Salas, A. I., Lopez, F. L., De Lecea, L., Gahete, M. D., Castano, J. P., Luque, R. M. 2015; 156 (6): 1958-1964
  • Obesity Alters Gene Expression for GH/IGF-I Axis in Mouse Mammary Fat Pads: Differential Role of Cortistatin and Somatostatin PLOS ONE Villa-Osaba, A., Gahete, M. D., Cordoba-Chacon, J., de Lecea, L., Pozo-Salas, A. I., Javier Delgado-Lista, F., Alvarez-Benito, M., Lopez-Miranda, J., Luque, R. M., Castano, J. P. 2015; 10 (3)

    Abstract

    Locally produced growth hormone (GH) and IGF-I are key factors in the regulation of mammary gland (MG) development and may be important in breast cancer development/progression. Somatostatin (SST) and cortistatin (CORT) regulate GH/IGF-I axis at various levels, but their role in regulating GH/IGF-I in MGs remains unknown. Since obesity alters the expression of these systems in different tissues and is associated to MG (patho) physiology, we sought to investigate the role of SST/CORT in regulating GH/IGF-I system in the MGs of lean and obese mice. Therefore, we analyzed GH/IGF-I as well as SST/CORT and ghrelin systems expression in the mammary fat pads (MFPs) of SST- or CORT-knockout (KO) mice and their respective littermate-controls fed a low-fat (LF) or a high-fat (HF) diet for 16wks. Our results demonstrate that the majority of the components of GH/IGF-I, SST/CORT and ghrelin systems are locally expressed in mouse MFP. Expression of elements of the GH/IGF-I axis was significantly increased in MFPs of HF-fed control mice while lack of endogenous SST partially suppressed, and lack of CORT completely blunted, the up-regulation observed in obese WT-controls. Since SST/CORT are known to exert an inhibitory role on the GH/IGFI axis, the increase in SST/CORT-receptor sst2 expression in MFPs of HF-fed CORT- and SST-KOs together with an elevation on circulating SST in CORT-KOs could explain the differences observed. These results offer new information on the factors (GH/IGF-I axis) involved in the endocrine/metabolic dysregulation of MFPs in obesity, and suggest that CORT is not a mere SST sibling in regulating MG physiology.

    View details for DOI 10.1371/journal.pone.0120955

    View details for Web of Science ID 000351880000116

    View details for PubMedID 25806796

    View details for PubMedCentralID PMC4373840

  • Antagonistic interplay between hypocretin and leptin in the lateral hypothalamus regulates stress responses NATURE COMMUNICATIONS Bonnavion, P., Jackson, A. C., Carter, M. E., de Lecea, L. 2015; 6

    Abstract

    The hypothalamic-pituitary-adrenal (HPA) axis functions to coordinate behavioural and physiological responses to stress in a manner that depends on the behavioural state of the organism. However, the mechanisms through which arousal and metabolic states influence the HPA axis are poorly understood. Here using optogenetic approaches in mice, we show that neurons that produce hypocretin (Hcrt)/orexin in the lateral hypothalamic area (LHA) regulate corticosterone release and a variety of behaviours and physiological hallmarks of the stress response. Interestingly, we found that Hcrt neuronal activity and Hcrt-mediated stress responses were inhibited by the satiety hormone leptin, which acts, in part, through a network of leptin-sensitive neurons in the LHA. These data demonstrate how peripheral metabolic signals interact with hypothalamic neurons to coordinate stress and arousal and suggest one mechanism through which hyperarousal or altered metabolic states may be linked with abnormal stress responses.

    View details for DOI 10.1038/ncomms7266

    View details for Web of Science ID 000350202800016

    View details for PubMedID 25695914

    View details for PubMedCentralID PMC4335349

  • The Hypocretin/Orexin System: An Increasingly Important Role in Neuropsychiatry MEDICINAL RESEARCH REVIEWS Chen, Q., de Lecea, L., Hu, Z., Gao, D. 2015; 35 (1): 152-197

    Abstract

    Hypocretins, also named as orexins, are excitatory neuropeptides secreted by neurons specifically located in lateral hypothalamus and perifornical areas. Orexinergic fibers are extensively distributed in various brain regions and involved in a number of physiological functions, such as arousal, cognition, stress, appetite, and metabolism. Arousal is the most important function of orexin system as dysfunction of orexin signaling leads to narcolepsy. In addition to narcolepsy, orexin dysfunction is associated with serious neural disorders, including addiction, depression, and anxiety. However, some results linking orexin with these disorders are still contradictory, which may result from differences of detection methods or the precision of tools used in measurements; strategies targeted to orexin system (e.g., antagonists to orexin receptors, gene delivery, and cell transplantation) are promising new tools for treatment of neuropsychiatric disorders, though studies are still in a stage of preclinical or clinical research.

    View details for DOI 10.1002/med.21326

    View details for Web of Science ID 000346080200005

    View details for PubMedID 25044006

  • Optogenetic Dissection of Neural Circuit Function in Behaving Animals NEURAL TRACING METHODS: TRACING NEURONS AND THEIR CONNECTIONS Herrera, C., Adamantidis, A., Zhang, F., Deisseroth, K., de Lecea, L., Arenkiel, B. R. 2015; 92: 143–60
  • Optogenetic control of hypocretin (orexin) neurons and arousal circuits. Current topics in behavioral neurosciences de Lecea, L. n. 2015; 25: 367–78

    Abstract

    In 1998, our group discovered a cDNA that encoded the precursor of two putative neuropeptides that we called hypocretins for their hypothalamic expression and their similarity to the secretin family of neuropeptides. In the last 16 years, numerous studies have placed the hypocretin system as an integrator of homeostatic functions with a crucial, non-redundant function as arousal stabilizer. We recently applied optogenetic methods to interrogate the role of individual neuronal circuits in sleep-to-wake transitions. The neuronal connections between the hypocretin system and the locus coeruleus (LC) seem to be crucial in establishing the appropriate dynamic of spontaneous awakenings.

    View details for PubMedID 25502546

    View details for PubMedCentralID PMC5047484

  • Optogenetics in Freely Moving Mammals: Dopamine and Reward. Cold Spring Harbor protocols Zhang, F., Tsai, H., Airan, R. D., Stuber, G. D., Adamantidis, A. R., de Lecea, L., Bonci, A., Deisseroth, K. 2015; 2015 (8): pdb top086330-?

    Abstract

    Brain reward systems play a central role in the cognitive and hedonic behaviors of mammals. Multiple neuron types and brain regions are involved in reward processing, posing fascinating scientific questions, and major experimental challenges. Using diverse approaches including genetics, electrophysiology, imaging, and behavioral analysis, a large body of research has focused on both normal functioning of the reward circuitry and on its potential significance in neuropsychiatric diseases. In this introduction, we illustrate a real-world application of optogenetics to mammalian behavior and physiology, delineating procedures and technologies for optogenetic control of individual components of the reward circuitry. We describe the experimental setup and protocol for integrating optogenetic modulation of dopamine neurons with fast-scan cyclic voltammetry, conditioned place preference, and operant conditioning to assess the causal role of well-defined electrical and biochemical signals in reward-related behavior.

    View details for DOI 10.1101/pdb.top086330

    View details for PubMedID 26240415

  • A Framework for Quantitative Modeling of Neural Circuits Involved in Sleep-to-Wake Transition. Frontiers in neurology Sorooshyari, S., Huerta, R., de Lecea, L. 2015; 6: 32-?

    Abstract

    Identifying the neuronal circuits and dynamics of sleep-to-wake transition is essential to understanding brain regulation of behavioral states, including sleep-wake cycles, arousal, and hyperarousal. Recent work by different laboratories has used optogenetics to determine the role of individual neuromodulators in state transitions. The optogenetically driven data do not yet provide a multi-dimensional schematic of the mechanisms underlying changes in vigilance states. This work presents a modeling framework to interpret, assist, and drive research on the sleep-regulatory network. We identify feedback, redundancy, and gating hierarchy as three fundamental aspects of this model. The presented model is expected to expand as additional data on the contribution of each transmitter to a vigilance state becomes available. Incorporation of conductance-based models of neuronal ensembles into this model and existing models of cortical excitability will provide more comprehensive insight into sleep dynamics as well as sleep and arousal-related disorders.

    View details for DOI 10.3389/fneur.2015.00032

    View details for PubMedID 25767461

    View details for PubMedCentralID PMC4341569

  • Potential role of orexin and sleep modulation in the pathogenesis of Alzheimer's disease JOURNAL OF EXPERIMENTAL MEDICINE Roh, J. H., Jiang, H., Finn, M. B., Stewart, F. R., Mahan, T. E., Cirrito, J. R., Heda, A., Snider, B. J., Li, M., Yanagisawa, M., de Lecea, L., Holtzman, D. M. 2014; 211 (13): 2487-2496

    Abstract

    Age-related aggregation of amyloid-β (Aβ) is an upstream pathological event in Alzheimer's disease (AD) pathogenesis, and it disrupts the sleep-wake cycle. The amount of sleep declines with aging and to a greater extent in AD. Poor sleep quality and insufficient amounts of sleep have been noted in humans with preclinical evidence of AD. However, how the amount and quality of sleep affects Aβ aggregation is not yet well understood. Orexins (hypocretins) initiate and maintain wakefulness, and loss of orexin-producing neurons causes narcolepsy. We tried to determine whether orexin release or secondary changes in sleep via orexin modulation affect Aβ pathology. Amyloid precursor protein (APP)/Presenilin 1 (PS1) transgenic mice, in which the orexin gene is knocked out, showed a marked decrease in the amount of Aβ pathology in the brain with an increase in sleep time. Focal overexpression of orexin in the hippocampus in APP/PS1 mice did not alter the total amount of sleep/wakefulness and the amount of Aβ pathology. In contrast, sleep deprivation or increasing wakefulness by rescue of orexinergic neurons in APP/PS1 mice lacking orexin increased the amount of Aβ pathology in the brain. Collectively, modulation of orexin and its effects on sleep appear to modulate Aβ pathology in the brain.

    View details for DOI 10.1084/jem.20141788

    View details for Web of Science ID 000346366100001

    View details for PubMedID 25422493

    View details for PubMedCentralID PMC4267230

  • Hypocretin (orexin) neuromodulation of stress and reward pathways CURRENT OPINION IN NEUROBIOLOGY Giardino, W. J., de Lecea, L. 2014; 29: 103-108

    Abstract

    Hypocretin (also known as orexin) is a peptide neuromodulator that is expressed exclusively in the lateral hypothalamic area and plays a fundamental role in wakefulness and arousal. Chronic stress and compulsive drug-seeking are two examples of dysregulated states of hyperarousal that are influenced by hypocretin transmission throughout hypothalamic, extended amygdala, brainstem, and mesolimbic pathways. Here, we review current advances in the understanding of hypocretin's modulatory actions underlying conditions of negative and positive emotional valence, focusing particularly on mechanisms that facilitate adaptive (and maladaptive) responses to stressful or rewarding environmental stimuli. We conclude by discussing progress toward integrated theories for hypocretin modulation of divergent behavioral domains.

    View details for DOI 10.1016/j.conb.2014.07.006

    View details for Web of Science ID 000347128200015

    View details for PubMedID 25050887

    View details for PubMedCentralID PMC4267967

  • Basal Forebrain Cholinergic Modulation of Sleep Transitions SLEEP Irmak, S. O., de Lecea, L. 2014; 37 (12): 1941-U104

    Abstract

    The basal forebrain cholinergic system is involved in cognitive processes that require an attentive state, an increased level of arousal, and/ or cortical activation associated with low amplitude fast EEG activity. The activity of most neurons in the basal forebrain cholinergic space is tightly correlated with the cortical EEG and the activity state. While most cholinergic neurons fire maximally during waking and REM sleep, the activity of other types of basal forebrain neurons vastly differs across different arousal and sleep states. Numerous studies have suggested a role for the basal forebrain cholinergic neurons in eliciting cortical activation and arousal. However, the intricate local connectivity within the region requires the use of cell-specific manipulation methods to demonstrate such a causal relationship.Here we have combined optogenetics with surface EEG recordings in freely moving mice in order to investigate the effects of acute cholinergic activation on the dynamics of sleep-to-wake transitions. We recorded from naturally sleeping animals and analyzed transitions from NREM sleep to REM sleep and/ or wakefulness in response to photo-stimulation of cholinergic neurons in substantia innominata.Our results show that optogenetic activation of BF cholinergic neurons during NREM sleep is sufficient to elicit cortical activation and facilitate state transitions, particularly transitions to wakefulness and arousal, at a time scale similar to the activation induced by other subcortical systems. Our results provide in vivo cell-specific demonstration for the role of basal forebrain cholinergic system in induction of wakefulness and arousal.

    View details for DOI 10.5665/sleep.4246

    View details for Web of Science ID 000345827600011

    View details for PubMedID 25325504

  • Control of sleep-to-wake transitions via fast amino acid and slow neuropeptide transmission NEW JOURNAL OF PHYSICS Mosqueiro, T., de Lecea, L., Huerta, R. 2014; 16
  • Light and chemical control of neuronal circuits: possible applications in neurotherapy. Expert review of neurotherapeutics Whittle, A. J., Walsh, J., de Lecea, L. 2014; 14 (9): 1007-1017

    Abstract

    Millions of people worldwide suffer from diseases that result from a failure of central pathways to regulate behavioral and physiological processes. Advances in genetics and pharmacology have already allowed us to appreciate that rather than this dysregulation being systemic throughout the brain, it is usually rooted in specific subsets of dysfunctional cells within discrete neurological circuits. This article discusses the advent of opto- and chemogenetic tools and how they are providing the means to dissect these circuits with a degree of temporal and spatial sensitivity not previously possible. We also highlight the potential applications for treating disease and the key developments likely to have the greatest impact over the next 5 years.

    View details for DOI 10.1586/14737175.2014.948850

    View details for PubMedID 25115180

  • Optogenetic control of hypocretin/orexin neurons de Lecea, L. AMER CHEMICAL SOC. 2014
  • Lack of Endogenous Cortistatin but Not Somatostatin Exacerbates Carcinogen-Induced Mammary Gland Tumorigenesis in Mice Gahete, M. D., Villa-Osaba, A., Cordoba-Chacon, J., de Lecea, L., Gracia-Navarro, F., Gonzalez-Rey, E., Luque, R. M., Casta, J. P. ENDOCRINE SOC. 2014
  • Cortistatin Is a Key Factor Regulating the Gender-Dependent Response of the Growth Hormone (GH) and Adrenocorticotropin (ACTH) Axes to Fasting in Mice Cordoba-Chacon, J., Gahete, M. D., Pozo-Salas, A., de Lecea, L., Castaao, J. P., Luque, R. M. ENDOCRINE SOC. 2014
  • Hypocretin (orexin) regulation of sleep-to-wake transitions FRONTIERS IN PHARMACOLOGY de Lecea, L., Huerta, R. 2014; 5

    Abstract

    The hypocretin (Hcrt), also known as orexin, peptides are essential for arousal stability. Here we discuss background information about the interaction of Hcrt with other neuromodulators, including norepinephrine and acetylcholine probed with optogenetics. We conclude that Hcrt neurons integrate metabolic, circadian and limbic inputs and convey this information to a network of neuromodulators, each of which has a different role on the dynamic of sleep-to-wake transitions. This model may prove useful to predict the effects of orexin receptor antagonists in sleep disorders and other conditions.

    View details for DOI 10.3389/fphar.2014.00016

    View details for Web of Science ID 000347042700001

    View details for PubMedID 24575043

    View details for PubMedCentralID PMC3921570

  • Establishing a fiber-optic-based optical neural interface. Cold Spring Harbor protocols Adamantidis, A. R., Zhang, F., de Lecea, L., Deisseroth, K. 2014; 2014 (8): pdb prot083337-?

    Abstract

    Selective expression of opsins in genetically defined neurons makes it possible to control a subset of neurons without affecting nearby cells and processes in the intact brain, but light must still be delivered to the target brain structure. Light scattering limits the delivery of light from the surface of the brain. For this reason, we have developed a fiber-optic-based optical neural interface (ONI), which allows optical access to any brain structure in freely moving mammals. The ONI system is constructed by modifying the small animal cannula system from PlasticsOne. The system for bilateral stimulation consists of a bilateral cannula guide that has been stereotactically implanted over the target brain region, a screw cap for securing the optical fiber to the animal's head, a fiber guard modified from the internal cannula adapter, and a bare fiber whose length is customized based on the depth of the target region. For unilateral stimulation, a single-fiber system can be constructed using unilateral cannula parts from PlasticsOne. We describe here the preparation of the bilateral ONI system and its use in optical stimulation of the mouse or rat brain. Delivery of opsin-expressing virus and implantation of the ONI may be conducted in the same surgical session; alternatively, with a transgenic animal no opsin virus is delivered during the surgery. Similar procedures are useful for deep or superficial injections (even for neocortical targets, although in some cases surface light-emitting diodes or cortex-apposed fibers can be used for the most superficial cortical targets).

    View details for DOI 10.1101/pdb.prot083337

    View details for PubMedID 25086020

  • Control of sleep-to-wake transitions via fast aminoacid and slow neuropeptide transmission. New journal of physics Mosqueiro, T. n., de Lecea, L. n., Huerta, R. n. 2014; 16

    Abstract

    The Locus Coeruleus (LC) modulates cortical, subcortical, cerebellar, brainstem and spinal cord circuits and it expresses receptors for neuromodulators that operate in a time scale of several seconds. Evidences from anatomical, electrophysiological and optogenetic experiments have shown that LC neurons receive input from a group of neurons called Hypocretins (HCRTs) that release a neuropeptide called hypocretin. It is less known how these two groups of neurons can be coregulated using GABAergic neurons. Since the time scales of GABA A inhibition is several orders of magnitude faster than the hypocretin neuropeptide effect, we investigate the limits of circuit activity regulation using a realistic model of neurons. Our investigation shows that GABA A inhibition is insufficient to control the activity levels of the LCs. Despite slower forms of GABA A can in principle work, there is not much plausibility due to the low probability of the presence of slow GABA A and lack of robust stability at the maximum firing frequencies. The best possible control mechanism predicted by our modeling analysis is the presence of inhibitory neuropeptides that exert effects in a similar time scale as the hypocretin/orexin. Although the nature of these inhibitory neuropeptides has not been identified yet, it provides the most efficient mechanism in the modeling analysis. Finally, we present a reduced mean-field model that perfectly captures the dynamics and the phenomena generated by this circuit. This investigation shows that brain communication involving multiple time scales can be better controlled by employing orthogonal mechanisms of neural transmission to decrease interference between cognitive processes and hypothalamic functions.

    View details for PubMedID 25598695

    View details for PubMedCentralID PMC4292803

  • The hypocretins/orexins: integrators of multiple physiological functions BRITISH JOURNAL OF PHARMACOLOGY Li, J., Hu, Z., de Lecea, L. 2014; 171 (2): 332-350

    Abstract

    The hypocretins (Hcrts), also known as orexins, are two peptides derived from a single precursor produced in the posterior lateral hypothalamus. Over the past decade, the orexin system has been associated with numerous physiological functions, including sleep/arousal, energy homeostasis, endocrine, visceral functions and pathological states, such as narcolepsy and drug abuse. Here, we review the discovery of Hcrt/orexins and their receptors and propose a hypothesis as to how the orexin system orchestrates these multifaceted physiological functions.

    View details for DOI 10.1111/bph.12415

    View details for Web of Science ID 000328712000005

    View details for PubMedID 24102345

  • Optogenetics: opsins and optical interfaces in neuroscience. Cold Spring Harbor protocols Adamantidis, A. R., Zhang, F., de Lecea, L., Deisseroth, K. 2014; 2014 (8): pdb top083329-?

    Abstract

    Optogenetics is defined as the integration of optics and genetics to control well-defined events within specified cells of living tissue. In this introduction, we focus on the basic techniques necessary for employing microbial opsins as optogenetic tools in mammalian brains. We provide a guide for the fundamentals of optogenetic application-selecting an opsin, implementing expression of opsins based on the neuroscientific experimental requirements, and adapting the corresponding optical hardware for delivery of light into mammalian brains.

    View details for DOI 10.1101/pdb.top083329

    View details for PubMedID 25086025

  • Orexin/hypocretin system modulates amygdala-dependent threat learning through the locus coeruleus PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Sears, R. M., Fink, A. E., Wigestrand, M. B., Farb, C. R., de Lecea, L., LeDoux, J. E. 2013; 110 (50): 20260-20265

    Abstract

    Survival in a dangerous environment requires learning about stimuli that predict harm. Although recent work has focused on the amygdala as the locus of aversive memory formation, the hypothalamus has long been implicated in emotional regulation, and the hypothalamic neuropeptide orexin (hypocretin) is involved in anxiety states and arousal. Nevertheless, little is known about the role of orexin in aversive memory formation. Using a combination of behavioral pharmacology, slice physiology, and optogenetic techniques, we show that orexin acts upstream of the amygdala via the noradrenergic locus coeruleus to enable threat (fear) learning, specifically during the aversive event. Our results are consistent with clinical studies linking orexin levels to aversive learning and anxiety in humans and dysregulation of the orexin system may contribute to the etiology of fear and anxiety disorders.

    View details for DOI 10.1073/pnas.1320325110

    View details for Web of Science ID 000328061700069

    View details for PubMedID 24277819

    View details for PubMedCentralID PMC3864341

  • Sleep to forget: interference of fear memories during sleep. Molecular psychiatry Rolls, A., Makam, M., Kroeger, D., Colas, D., De Lecea, L., Heller, H. C. 2013; 18 (11): 1166-1170

    Abstract

    Memories are consolidated and strengthened during sleep. Here we show that memories can also be weakened during sleep. We used a fear-conditioning paradigm in mice to condition footshock to an odor (conditioned stimulus (CS)). Twenty-four hours later, presentation of the CS odor during sleep resulted in an enhanced fear response when tested during subsequent wake. However, if the re-exposure of the CS odor during sleep was preceded by bilateral microinjections of a protein synthesis inhibitor into the basolateral amygdala, the subsequent fear response was attenuated. These findings demonstrate that specific fear memories can be selectively reactivated and either strengthened or attenuated during sleep, suggesting the potential for developing sleep therapies for emotional disorders.

    View details for DOI 10.1038/mp.2013.121

    View details for PubMedID 24081009

    View details for PubMedCentralID PMC5036945

  • Paradoxical Effect of Cortistatin Treatment and Its Deficiency on Experimental Autoimmune Encephalomyelitis JOURNAL OF IMMUNOLOGY Souza-Moreira, L., Morell, M., Delgado-Maroto, V., Pedreno, M., Martinez-Escudero, L., Caro, M., O'Valle, F., Luque, R., Gallo, M., de Lecea, L., Castano, J. P., Gonzalez-Rey, E. 2013; 191 (5): 2144-2154

    Abstract

    Cortistatin is a cyclic-neuropeptide produced by brain cortex and immune cells that shows potent anti-inflammatory activity. In this article, we investigated the effect of cortistatin in two models of experimental autoimmune encephalomyelitis (EAE) that mirror chronic and relapsing-remitting multiple sclerosis. A short-term systemic treatment with cortistatin reduced clinical severity and incidence of EAE, the appearance of inflammatory infiltrates in spinal cord, and the subsequent demyelination and axonal damage. This effect was associated with a reduction of the two deleterious components of the disease, namely, the autoimmune and inflammatory response. Cortistatin decreased the presence/activation of encephalitogenic Th1 and Th17 cells in periphery and nervous system, and downregulated various inflammatory mediators, whereas it increased the number of regulatory T cells with suppressive effects on the encephalitogenic response. Moreover, cortistatin regulated glial activity and favored an active program of neuroprotection/regeneration. We further used cortistatin-deficient mice to investigate the role of endogenous cortistatin in the control of immune responses. Surprisingly, cortistatin-deficient mice were partially resistant to EAE and other inflammatory disorders, despite showing competent inflammatory/autoreactive responses. This unexpected phenotype was associated with elevated circulating glucocorticoids and an anxiety-like behavior. Our findings provide a powerful rationale for the assessment of the efficacy of cortistatin as a novel multimodal therapeutic approach to treat multiple sclerosis and identify cortistatin as a key endogenous component of neuroimmune system.

    View details for DOI 10.4049/jimmunol.1300384

    View details for Web of Science ID 000323393300015

    View details for PubMedID 23918980

  • Optogenetics in psychiatric diseases. Current opinion in neurobiology Touriño, C., Eban-Rothschild, A., de Lecea, L. 2013; 23 (3): 430-435

    Abstract

    Optogenetic tools have revolutionized the field of neuroscience, and brought the study of neural circuits to a higher level. Optogenetics has significantly improved our understanding not only of the neuronal connections and function of the healthy brain, but also of the neuronal changes that lead to psychiatric disorders. In this review, we summarize recent optogenetic studies that explored different brain circuits involved in natural behaviors, such as sleep and arousal, reward, fear, and social and aggressive behavior. In addition, we describe how alterations in these circuits may lead to psychiatric disorders such as addiction, anxiety, depression, or schizophrenia.

    View details for DOI 10.1016/j.conb.2013.03.007

    View details for PubMedID 23642859

  • Cortistatin Inhibits Migration and Proliferation of Human Vascular Smooth Muscle Cells and Decreases Neointimal Formation on Carotid Artery Ligation CIRCULATION RESEARCH Duran-Prado, M., Morell, M., Delgado-Maroto, V., Castano, J. P., Aneiros-Fernandez, J., de Lecea, L., Culler, M. D., Hernandez-Cortes, P., O'Valle, F., Delgado, M. 2013; 112 (11): 1444-?

    Abstract

    Proliferation and migration of smooth muscle cells (SMCs) are key steps for the progression of atherosclerosis and restenosis. Cortistatin is a multifunctional neuropeptide belonging to the somatostatin family that exerts unique functions in the nervous and immune systems. Cortistatin is elevated in plasma of patients experiencing coronary heart disease and attenuates vascular calcification.To investigate the occurrence of vascular cortistatin and its effects on the proliferation and migration of SMCs in vitro and in vivo and to delimitate the receptors and signal transduction pathways governing its actions.SMCs from mouse carotid and human aortic arteries and from human atherosclerotic plaques highly expressed cortistatin. Cortistatin expression positively correlated with the progression of arterial intima hyperplasia. Cortistatin inhibited platelet-derived growth factor-stimulated proliferation of human aortic SMCs via binding to somatostatin receptors (sst2 and sst5) and ghrelin receptor, induction of cAMP and p38-mitogen-activated protein kinase, and inhibition of Akt activity. Moreover, cortistatin impaired lamellipodia formation and migration of human aortic SMCs toward platelet-derived growth factor by inhibiting, in a ghrelin-receptor-dependent manner, Rac1 activation and cytosolic calcium increases. These effects on SMC proliferation and migration correlated with an inhibitory action of cortistatin on the neointimal formation in 2 models of carotid arterial ligation. Endogenous cortistatin seems to play a critical role in regulating SMC function because cortistatin-deficient mice developed higher neointimal hyperplasic lesions than wild-type mice.Cortistatin emerges as a natural endogenous regulator of SMCs under pathological conditions and an attractive candidate for the pharmacological management of vascular diseases that course with neointimal lesion formation.

    View details for DOI 10.1161/CIRCRESAHA.112.300695

    View details for Web of Science ID 000319448900013

    View details for PubMedID 23595952

  • Functional wiring of hypocretin and LC-NE neurons: implications for arousal FRONTIERS IN BEHAVIORAL NEUROSCIENCE Carter, M. E., de Lecea, L., Adamantidis, A. 2013; 7

    Abstract

    To survive in a rapidly changing environment, animals must sense their external world and internal physiological state and properly regulate levels of arousal. Levels of arousal that are abnormally high may result in inefficient use of internal energy stores and unfocused attention to salient environmental stimuli. Alternatively, levels of arousal that are abnormally low may result in the inability to properly seek food, water, sexual partners, and other factors necessary for life. In the brain, neurons that express hypocretin neuropeptides may be uniquely posed to sense the external and internal state of the animal and tune arousal state according to behavioral needs. In recent years, we have applied temporally precise optogenetic techniques to study the role of these neurons and their downstream connections in regulating arousal. In particular, we have found that noradrenergic neurons in the brainstem locus coeruleus (LC) are particularly important for mediating the effects of hypocretin neurons on arousal. Here, we discuss our recent results and consider the implications of the anatomical connectivity of these neurons in regulating the arousal state of an organism across various states of sleep and wakefulness.

    View details for DOI 10.3389/fnbeh.2013.00043

    View details for Web of Science ID 000319055000001

    View details for PubMedID 23730276

    View details for PubMedCentralID PMC3657625

  • Hypothalamic Neurotensin Projections Promote Reward by Enhancing Glutamate Transmission in the VTA JOURNAL OF NEUROSCIENCE Kempadoo, K. A., Tourino, C., Cho, S. L., Magnani, F., Leinninger, G., Stuber, G. D., Zhang, F., Myers, M. G., Deisseroth, K., de Lecea, L., Bonci, A. 2013; 33 (18): 7618-?

    Abstract

    The lateral hypothalamus (LH) sends a dense glutamatergic and peptidergic projection to dopamine neurons in the ventral tegmental area (VTA), a cell group known to promote reinforcement and aspects of reward. The role of the LH to VTA projection in reward-seeking behavior can be informed by using optogenetic techniques to dissociate the actions of LH neurons from those of other descending forebrain inputs to the VTA. In the present study, we identify the effect of neurotensin (NT), one of the most abundant peptides in the LH to VTA projection, on excitatory synaptic transmission in the VTA and reward-seeking behavior. Mice displayed robust intracranial self-stimulation of LH to VTA fibers, an operant behavior mediated by NT 1 receptors (Nts1) and NMDA receptors. Whole-cell patch-clamp recordings of VTA dopamine neurons demonstrated that NT (10 nm) potentiated NMDA-mediated EPSCs via Nts1. Results suggest that NT release from the LH into the VTA activates Nts1, thereby potentiating NMDA-mediated EPSCs and promoting reward. The striking behavioral and electrophysiological effects of NT and glutamate highlight the LH to VTA pathway as an important component of reward.

    View details for DOI 10.1523/JNEUROSCI.2588-12.2013

    View details for Web of Science ID 000318420400002

    View details for PubMedID 23637156

  • Repeated in vivo exposure of cocaine induces long-lasting synaptic plasticity in hypocretin/orexin-producing neurons in the lateral hypothalamus in mice JOURNAL OF PHYSIOLOGY-LONDON Rao, Y., Mineur, Y. S., Gan, G., Wang, A. H., Liu, Z., Wu, X., Suyama, S., de Lecea, L., Horvath, T. L., Picciotto, M. R., Gao, X. 2013; 591 (7): 1951-1966

    Abstract

    Hypocretin (orexin), a neuropeptide synthesized exclusively in the perifornical/lateral hypothalamus, is critical for drug seeking and relapse, but it is not clear how the circuitry centred on hypocretin-producing neurons (hypocretin neurons) is modified by drugs of abuse and how changes in this circuit might alter behaviours related to drug addiction. In this study, we show that repeated, but not single, in vivo cocaine administration leads to a long-lasting, experience-dependent potentiation of glutamatergic synapses on hypocretin neurons in mice following a cocaine-conditioned place preference (CPP) protocol. The synaptic potentiation occurs postsynaptically and probably involves up-regulation of AMPA-type glutamate receptors on hypocretin neurons. Phosphorylation of cAMP response element-binding protein (CREB) is also significantly increased in hypocretin neurons in cocaine-treated animals, suggesting that CREB-mediated pathways may contribute to synaptic potentiation in these cells. Furthermore, the potentiation of synaptic efficacy in hypocretin neurons persists during cocaine withdrawal, but reverses to baseline levels after prolonged abstinence. Finally, the induction of long-term potentiation (LTP) triggered by a high-frequency stimulation is facilitated in hypocretin neurons in cocaine-treated mice, suggesting that long-lasting changes in synapses onto hypocretin neurons would probably be further potentiated by other stimuli (such as concurrent environmental cues) paired with the drug. In summary, we show here that hypocretin neurons undergo experience-dependent synaptic potentiation that is distinct from that reported in other reward systems, such as the ventral tegmental area, following exposure to cocaine. These findings support the idea that the hypocretin system is important for behavioural changes associated with cocaine administration in animals and humans.

    View details for DOI 10.1113/jphysiol.2012.246983

    View details for Web of Science ID 000316918300030

    View details for PubMedID 23318871

    View details for PubMedCentralID PMC3624862