Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 21(11): e3002389, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37983289

RESUMEN

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 LTß 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.


Asunto(s)
Trypanosoma brucei brucei , Tripanosomiasis Africana , Humanos , Animales , Ratones , Infección Persistente , Meninges/metabolismo , Tejido Linfoide/metabolismo , Autoanticuerpos
2.
Cell Mol Life Sci ; 81(1): 288, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38970689

RESUMEN

Orexinergic neurons are critically involved in regulating arousal, wakefulness, and appetite. Their dysfunction has been associated with sleeping disorders, and non-peptide drugs are currently being developed to treat insomnia and narcolepsy. Yet, no light-regulated agents are available to reversibly control their activity. To meet this need, a photoswitchable peptide analogue of the endogenous neuroexcitatory peptide orexin-B was designed, synthesized, and tested in vitro and in vivo. This compound - photorexin - is the first photo-reversible ligand reported for orexin receptors. It allows dynamic control of activity in vitro (including almost the same efficacy as orexin-B, high nanomolar potency, and subtype selectivity to human OX2 receptors) and in vivo in zebrafish larvae by direct application in water. Photorexin induces dose- and light-dependent changes in locomotion and a reduction in the successive induction reflex that is associated with sleep behavior. Molecular dynamics calculations indicate that trans and cis photorexin adopt similar bent conformations and that the only discriminant between their structures and activities is the positioning of the N-terminus. This, in the case of the more active trans isomer, points towards the OX2 N-terminus and extra-cellular loop 2, a region of the receptor known to be involved in ligand binding and recognition consistent with a "message-address" system. Thus, our approach could be extended to several important families of endogenous peptides, such as endothelins, nociceptin, and dynorphins among others, that bind to their cognate receptors through a similar mechanism: a "message" domain involved in receptor activation and signal transduction, and an "address" sequence for receptor occupation and improved binding affinity.


Asunto(s)
Luz , Receptores de Orexina , Orexinas , Pez Cebra , Receptores de Orexina/metabolismo , Receptores de Orexina/química , Animales , Orexinas/metabolismo , Humanos , Locomoción/efectos de los fármacos , Simulación de Dinámica Molecular , Larva/metabolismo , Larva/efectos de los fármacos , Células HEK293 , Ligandos
3.
Proc Natl Acad Sci U S A ; 119(46): e2206828119, 2022 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-36343238

RESUMEN

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.


Asunto(s)
Epilepsia del Lóbulo Temporal , Epilepsia , Animales , Epilepsia/terapia , Encéfalo/diagnóstico por imagen , Encéfalo/fisiología , Ultrasonografía , Hipocampo/diagnóstico por imagen
4.
J Neuroinflammation ; 20(1): 48, 2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36829230

RESUMEN

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.


Asunto(s)
Envejecimiento Cognitivo , Microglía , Ratones , Animales , Microglía/metabolismo , Proteínas CLOCK/genética , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Plasticidad Neuronal
5.
J Intern Med ; 291(5): 533-556, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35043499

RESUMEN

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.


Asunto(s)
Narcolepsia , Neuropéptidos , Trastornos del Inicio y del Mantenimiento del Sueño , Humanos , Péptidos y Proteínas de Señalización Intracelular , Narcolepsia/tratamiento farmacológico , Neuropéptidos/fisiología , Orexinas
6.
J Neurosci ; 37(39): 9574-9592, 2017 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-28874450

RESUMEN

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.


Asunto(s)
Nivel de Alerta , Área Hipotalámica Lateral/fisiología , Neuronas/fisiología , Animales , Histamina/metabolismo , Área Hipotalámica Lateral/citología , Área Hipotalámica Lateral/metabolismo , Masculino , Potenciales de la Membrana , Ratones , Neuronas/clasificación , Sueño , Vigilia
7.
Cereb Cortex ; 26(4): 1590-1608, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25595181

RESUMEN

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.


Asunto(s)
Corteza Entorrinal/fisiología , Histamina/fisiología , Neuronas/fisiología , Aprendizaje Espacial/fisiología , Animales , Canales de Calcio/fisiología , Corteza Entorrinal/efectos de los fármacos , Ácido Glutámico/fisiología , Histamina/administración & dosificación , Masculino , Neuronas/efectos de los fármacos , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/fisiología , Ratas , Ratas Sprague-Dawley , Receptores Histamínicos H1/fisiología , Aprendizaje Espacial/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos , Ácido gamma-Aminobutírico/fisiología
8.
Int J Mol Sci ; 18(8)2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28809797

RESUMEN

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.


Asunto(s)
Nivel de Alerta/fisiología , Hipotálamo/fisiología , Red Nerviosa/fisiología , Neuronas/fisiología , Optogenética/métodos , Animales , Humanos , Hipotálamo/citología , Red Nerviosa/citología , Neuronas/citología
9.
J Physiol ; 594(22): 6443-6462, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27302606

RESUMEN

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.


Asunto(s)
Área Hipotalámica Lateral/metabolismo , Área Hipotalámica Lateral/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/fisiología , Humanos , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Neuropéptidos/metabolismo , Hormonas Hipofisarias/metabolismo
10.
Breast Cancer Res ; 18(1): 29, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26956474

RESUMEN

BACKGROUND: Somatostatin (SST) and cortistatin (CORT), two structurally and functionally related peptides, share a family of widespread receptors (sst1-5) to exert apparently similar biological actions, including endocrine/metabolic regulation and suppression of tumor cell proliferation. However, despite their therapeutic potential, attempts to apply SST-analogs to treat breast cancer have yielded unsatisfactory results. Actually, the specific roles of SST and CORT in mammary gland tumorigenesis (MGT), particularly in relation to metabolic dysregulation (i.e. obesity), remain unknown. METHODS: The role of endogenous SST and CORT in carcinogen-induced MGT was investigated under normal (lean) and obesity conditions. To that end, SST- and CORT-knockout (KO) mice and their respective littermate-controls, fed low-fat (LF) or high-fat (HF) diets, were treated with 7,12-dimethyl-benza-anthracene (DMBA) once a week (wk) for 3 wk, and MGT was monitored for 25 wk. Additionally, we examined the effect of SST or CORT removal in the development of the mammary gland. RESULTS: Lack of SST did not alter DMBA-induced MGT incidence under lean conditions; conversely, lack of endogenous CORT severely aggravated DMBA-induced MGT in LF-fed mice. These differences were not attributable to altered mammary gland development. HF-diet modestly increased the sensitivity to DMBA-induced carcinogenesis in control mice, whereas, as observed in LF-fed CORT-KO, HF-fed CORT-KO mice exhibited aggravated tumor incidence, discarding a major influence of obesity on these CORT actions. In marked contrast, HF-fed SST-KO mice exhibited much higher tumor incidence than LF-fed SST-KO mice, which could be associated with higher mammary complexity. CONCLUSIONS: Endogenous SST and CORT distinctly impact on DMBA-induced MGT, in a manner that is strongly dependent on the metabolic/endocrine milieu (lean vs. obese status). Importantly, CORT, rather than SST, could represent a major inhibitor of MGT under normal/lean-conditions, whereas both neuropeptides would similarly influence MGT under obesity conditions. The mechanisms mediating these different effects likely involve mammary development and hormones, but the precise underlying factors are still to be fully elucidated. However, our findings comprise suggestive evidence that CORT-like molecules, rather than classic SST-analogs, may help to identify novel tools for the medical treatment of breast cancer.


Asunto(s)
Carcinogénesis/genética , Neoplasias Mamarias Animales/genética , Neuropéptidos/genética , Somatostatina/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Femenino , Humanos , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/inducido químicamente , Neoplasias Mamarias Animales/patología , Ratones , Ratones Noqueados , Ratones Obesos/genética
11.
Curr Psychiatry Rep ; 18(1): 7, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26733323

RESUMEN

The hypocretins (Hcrts), also known as orexins, have been among the most intensely studied neuropeptide systems since their discovery about two decades ago. Anatomical evidence shows that the hypothalamic neurons that produce hypocretins/orexins project widely throughout the entire brain, innervating the noradrenergic locus coeruleus, the cholinergic basal forebrain, the dopaminergic ventral tegmental area, the serotonergic raphe nuclei, the histaminergic tuberomammillary nucleus, and many other brain regions. By interacting with other neural systems, the Hcrt system profoundly modulates versatile physiological processes including arousal, food intake, emotion, attention, and reward. Importantly, interruption of the interactions between these systems has the potential to cause neurological and psychiatric diseases. Here, we review the modulation of diverse neural systems by Hcrts and summarize potential therapeutic strategies based on our understanding of the Hcrt system's role in physiology and pathophysiological processes.


Asunto(s)
Trastornos Mentales , Sistema Nervioso , Orexinas/fisiología , Humanos , Hipotálamo/metabolismo , Trastornos Mentales/metabolismo , Trastornos Mentales/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso/metabolismo , Sistema Nervioso/fisiopatología , Neuropéptidos/fisiología
12.
Proc Natl Acad Sci U S A ; 110(50): 20260-5, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24277819

RESUMEN

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.


Asunto(s)
Amígdala del Cerebelo/fisiología , Miedo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Aprendizaje/fisiología , Locus Coeruleus/fisiología , Neuropéptidos/metabolismo , Estimulación Acústica , Animales , Benzoxazoles/administración & dosificación , Benzoxazoles/farmacología , Channelrhodopsins , Condicionamiento Clásico , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Masculino , Naftiridinas , Neuropéptidos/antagonistas & inhibidores , Optogenética , Orexinas , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Urea/administración & dosificación , Urea/análogos & derivados , Urea/farmacología
13.
Med Res Rev ; 35(1): 152-97, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25044006

RESUMEN

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.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/fisiología , Neuropéptidos/fisiología , Neuropsiquiatría , Humanos , Orexinas
14.
Circ Res ; 112(11): 1444-55, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23595952

RESUMEN

RATIONALE: 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. OBJECTIVE: 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. METHODS AND RESULTS: 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. CONCLUSIONS: 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.


Asunto(s)
Aterosclerosis/patología , Traumatismos de las Arterias Carótidas/patología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Neointima/patología , Neuropéptidos/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Humanos , Ligadura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neointima/tratamiento farmacológico , Neointima/metabolismo , Neuropéptidos/genética , Neuropéptidos/farmacología , ARN Mensajero/metabolismo , Receptores de Ghrelina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Somatostatina/metabolismo
15.
J Immunol ; 191(5): 2144-54, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23918980

RESUMEN

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.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Neuropéptidos/metabolismo , Linfocitos T/efectos de los fármacos , Animales , Encefalomielitis Autoinmune Experimental/patología , Femenino , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/inmunología , Médula Espinal/patología , Linfocitos T/inmunología
16.
Proc Natl Acad Sci U S A ; 109(39): E2635-44, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22955882

RESUMEN

Current models of sleep/wake regulation posit that Hypocretin (Hcrt)-expressing neurons in the lateral hypothalamus promote and stabilize wakefulness by projecting to subcortical arousal centers. However, the critical downstream effectors of Hcrt neurons are unknown. Here we use optogenetic, pharmacological, and computational tools to investigate the functional connectivity between Hcrt neurons and downstream noradrenergic neurons in the locus coeruleus (LC) during nonrapid eye movement (NREM) sleep. We found that photoinhibiting LC neurons during Hcrt stimulation blocked Hcrt-mediated sleep-to-wake transitions. In contrast, when LC neurons were optically stimulated to increase membrane excitability, concomitant photostimulation of Hcrt neurons significantly increased the probability of sleep-to-wake transitions compared with Hcrt stimulation alone. We also built a conductance-based computational model of Hcrt-LC circuitry that recapitulates our behavioral results using LC neurons as the main effectors of Hcrt signaling. These results establish the Hcrt-LC connection as a critical integrator-effector circuit that regulates NREM sleep/wake behavior during the inactive period. This coupling of distinct neuronal systems can be generalized to other hypothalamic integrator nuclei with downstream effector/output populations in the brain.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Locus Coeruleus/metabolismo , Neuropéptidos/metabolismo , Transducción de Señal/fisiología , Sueño REM/fisiología , Vigilia/fisiología , Neuronas Adrenérgicas/citología , Animales , Locus Coeruleus/citología , Ratones , Ratones Noqueados , Orexinas , Estimulación Luminosa
17.
J Neurosci ; 33(18): 7618-26, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23637156

RESUMEN

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.


Asunto(s)
Condicionamiento Operante/fisiología , Ácido Glutámico/metabolismo , Hipotálamo/fisiología , Neurotensina/metabolismo , Recompensa , Área Tegmental Ventral/fisiología , Animales , Proteínas Bacterianas/genética , Channelrhodopsins , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Hipotálamo/efectos de los fármacos , Técnicas In Vitro , Luz , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Vías Nerviosas/fisiología , Neurotensina/farmacología , Pirazoles/farmacología , Quinolinas/farmacología , Quinoxalinas/farmacología , Receptores de Neurotensina/antagonistas & inhibidores , Receptores de Neurotensina/deficiencia , Autoestimulación , Transducción de Señal/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo , Valina/análogos & derivados , Valina/farmacología , Área Tegmental Ventral/efectos de los fármacos
18.
Neurobiol Dis ; 63: 141-54, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24333694

RESUMEN

Clinical pain, as a consequence of inflammation or injury of peripheral organs (inflammatory pain) or nerve injury (neuropathic pain), represents a serious public health issue. Treatment of pain-related suffering requires knowledge of how pain signals are initially interpreted and subsequently transmitted and perpetuated. To limit duration and intensity of pain, inhibitory signals participate in pain perception. Cortistatin is a cyclic-neuropeptide that exerts potent inhibitory actions on cortical neurons and immune cells. Here, we found that cortistatin is a natural analgesic component of the peripheral nociceptive system produced by peptidergic nociceptive neurons of the dorsal root ganglia in response to inflammatory and noxious stimuli. Moreover, cortistatin is produced by GABAergic interneurons of deep layers of dorsal horn of spinal cord. By using cortistatin-deficient mice, we demonstrated that endogenous cortistatin critically tunes pain perception in physiological and pathological states. Furthermore, peripheral and spinal injection of cortistatin potently reduced nocifensive behavior, heat hyperalgesia and tactile allodynia in experimental models of clinical pain evoked by chronic inflammation, surgery and arthritis. The analgesic effects of cortistatin were independent of its anti-inflammatory activity and directly exerted on peripheral and central nociceptive terminals via Gαi-coupled somatostatin-receptors (mainly sstr2) and blocking intracellular signaling that drives neuronal plasticity including protein kinase A-, calcium- and Akt/ERK-mediated release of nociceptive peptides. Moreover, cortistatin could modulate, through its binding to ghrelin-receptor (GHSR1), pain-induced sensitization of secondary neurons in spinal cord. Therefore, cortistatin emerges as an anti-inflammatory factor with potent analgesic effects that offers a new approach to clinical pain therapy, especially in inflammatory states.


Asunto(s)
Analgésicos/uso terapéutico , Neuropéptidos/metabolismo , Dolor/tratamiento farmacológico , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Células Cultivadas , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Femenino , Ganglios Espinales/citología , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuropéptidos/deficiencia , Nitrobencenos/uso terapéutico , Dolor/etiología , Dolor/genética , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Médula Espinal/citología , Sulfonamidas/uso terapéutico , Factores de Tiempo
19.
Arthritis Rheum ; 65(5): 1390-401, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23371349

RESUMEN

OBJECTIVE: To investigate the role of the antiinflammatory neuropeptide cortistatin in chronic pain evoked by joint inflammation. METHODS: Thermal and mechanical hyperalgesia was evoked in mouse knee joints by intraplantar injection of tumor necrosis factor α and intraarticular infusion of Freund's complete adjuvant, and the analgesic effects of cortistatin, administered centrally, peripherally, and systemically, were assessed. In addition, the effects of cortistatin on the production of nociceptive peptides and the activation of pain signaling were assayed in dorsal root ganglion cultures and in inflammatory pain models. The role of endogenous cortistatin in pain sensitization and perpetuation of chronic inflammatory states was evaluated in cortistatin-deficient mice. Finally, the effect of noxious/inflammatory stimuli in the production of cortistatin by the peripheral nociceptive system was assayed in vitro and in vivo. RESULTS: Expression of cortistatin was observed in peptidergic nociceptors of the peripheral nociceptive system, and endogenous cortistatin was found to participate in the tuning of pain sensitization, especially in pathologic inflammatory conditions. Results showed that cortistatin acted both peripherally and centrally to reduce the tactile allodynia and heat hyperalgesia evoked by arthritis and peripheral tissue inflammation in mice, via mechanisms that were independent of its antiinflammatory action. These mechanisms involved direct action on nociceptive neurons and regulation of central sensitization. The analgesic effects of cortistatin in murine arthritic pain were linked to binding of the neuropeptide to somatostatin and ghrelin receptors, activation of the G protein subunit Gαi , impairment of ERK signaling, and decreased production of calcitonin gene-related peptide in primary nociceptors. CONCLUSION: These findings indicate that cortistatin is an antiinflammatory factor with potent analgesic effects that may offer a new approach to pain therapy in pathologic inflammatory states, including osteoarthritis and rheumatoid arthritis.


Asunto(s)
Analgesia , Artritis/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Neuropéptidos/farmacología , Dolor/tratamiento farmacológico , Animales , Artritis/inducido químicamente , Artritis/metabolismo , Péptido Relacionado con Gen de Calcitonina/metabolismo , Sensibilización del Sistema Nervioso Central , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Adyuvante de Freund/administración & dosificación , Adyuvante de Freund/toxicidad , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Ghrelina/metabolismo , Ghrelina/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Inyecciones Intraarticulares , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/deficiencia , Neuropéptidos/metabolismo , Dolor/inducido químicamente , Dolor/metabolismo , Umbral del Dolor , Unión Proteica , Receptores de Ghrelina/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Somatostatina/farmacología , Rodilla de Cuadrúpedos/efectos de los fármacos , Rodilla de Cuadrúpedos/metabolismo , Rodilla de Cuadrúpedos/fisiopatología , Factor de Necrosis Tumoral alfa/toxicidad
20.
Proc Natl Acad Sci U S A ; 108(32): 13305-10, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21788501

RESUMEN

Memory consolidation has been proposed as a function of sleep. However, sleep is a complex phenomenon characterized by several features including duration, intensity, and continuity. Sleep continuity is disrupted in different neurological and psychiatric conditions, many of which are accompanied by memory deficits. This finding has raised the question of whether the continuity of sleep is important for memory consolidation. However, current techniques used in sleep research cannot manipulate a single sleep feature while maintaining the others constant. Here, we introduce the use of optogenetics to investigate the role of sleep continuity in memory consolidation. We optogenetically targeted hypocretin/orexin neurons, which play a key role in arousal processes. We used optogenetics to activate these neurons at different intervals in behaving mice and were able to fragment sleep without affecting its overall amount or intensity. Fragmenting sleep after the learning phase of the novel object recognition (NOR) task significantly decreased the performance of mice on the subsequent day, but memory was unaffected if the average duration of sleep episodes was maintained at 62-73% of normal. These findings demonstrate the use of optogenetic activation of arousal-related nuclei as a way to systematically manipulate a specific feature of sleep. We conclude that regardless of the total amount of sleep or sleep intensity, a minimal unit of uninterrupted sleep is crucial for memory consolidation.


Asunto(s)
Memoria/fisiología , Privación de Sueño/fisiopatología , Sueño/genética , Sueño/efectos de la radiación , Animales , Electroencefalografía , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Memoria/efectos de la radiación , Ratones , Neuronas/metabolismo , Neuronas/patología , Neuronas/efectos de la radiación , Neuropéptidos/genética , Neuropéptidos/metabolismo , Orexinas , Estimulación Física , Privación de Sueño/complicaciones , Privación de Sueño/genética , Sueño REM/fisiología , Sueño REM/efectos de la radiación , Estrés Psicológico/complicaciones , Estrés Psicológico/fisiopatología , Análisis y Desempeño de Tareas , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA