Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 464
Filtrar
1.
Nat Commun ; 15(1): 4152, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755120

RESUMEN

Serotonin is a neuromodulator that affects multiple behavioral and cognitive functions. Nonetheless, how serotonin causes such a variety of effects via brain-wide projections and various receptors remains unclear. Here we measured brain-wide responses to optogenetic stimulation of serotonin neurons in the dorsal raphe nucleus (DRN) of the male mouse brain using functional MRI with an 11.7 T scanner and a cryoprobe. Transient activation of DRN serotonin neurons caused brain-wide activation, including the medial prefrontal cortex, the striatum, and the ventral tegmental area. The same stimulation under anesthesia with isoflurane decreased brain-wide activation, including the hippocampal complex. These brain-wide response patterns can be explained by DRN serotonergic projection topography and serotonin receptor expression profiles, with enhanced weights on 5-HT1 receptors. Together, these results provide insight into the DR serotonergic system, which is consistent with recent discoveries of its functions in adaptive behaviors.


Asunto(s)
Núcleo Dorsal del Rafe , Optogenética , Neuronas Serotoninérgicas , Serotonina , Animales , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/fisiología , Masculino , Neuronas Serotoninérgicas/metabolismo , Neuronas Serotoninérgicas/fisiología , Ratones , Serotonina/metabolismo , Imagen por Resonancia Magnética , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiología , Ratones Endogámicos C57BL , Encéfalo/metabolismo , Encéfalo/fisiología , Área Tegmental Ventral/fisiología , Área Tegmental Ventral/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiología , Receptores de Serotonina/metabolismo , Receptores de Serotonina/genética
2.
Sci Rep ; 14(1): 10190, 2024 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702366

RESUMEN

Dysfunction of central serotonergic neurons is known to cause depressive disorders in humans, who often show reproductive and/or glucose metabolism disorders. This study examined whether dorsal raphe (DR) serotonergic neurons sense high glucose availability to upregulate reproductive function via activating hypothalamic arcuate (ARC) kisspeptin neurons (= KNDy neurons), a dominant stimulator of gonadotropin-releasing hormone (GnRH)/gonadotropin pulses, using female rats and goats. RNA-seq and histological analysis revealed that stimulatory serotonin-2C receptor (5HT2CR) was mainly expressed in the KNDy neurons in female rats. The serotonergic reuptake inhibitor administration into the mediobasal hypothalamus (MBH), including the ARC, significantly blocked glucoprivic suppression of luteinizing hormone (LH) pulses and hyperglycemia induced by intravenous 2-deoxy-D-glucose (2DG) administration in female rats. A local infusion of glucose into the DR significantly increased in vivo serotonin release in the MBH and partly restored LH pulses and hyperglycemia in the 2DG-treated female rats. Furthermore, central administration of serotonin or a 5HT2CR agonist immediately evoked GnRH pulse generator activity, and central 5HT2CR antagonism blocked the serotonin-induced facilitation of GnRH pulse generator activity in ovariectomized goats. These results suggest that DR serotonergic neurons sense high glucose availability to reduce gluconeogenesis and upregulate reproductive function by activating GnRH/LH pulse generator activity in mammals.


Asunto(s)
Glucosa , Cabras , Hormona Liberadora de Gonadotropina , Hormona Luteinizante , Receptor de Serotonina 5-HT2C , Neuronas Serotoninérgicas , Animales , Hormona Luteinizante/metabolismo , Femenino , Receptor de Serotonina 5-HT2C/metabolismo , Ratas , Neuronas Serotoninérgicas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Glucosa/metabolismo , Serotonina/metabolismo , Kisspeptinas/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/efectos de los fármacos , Ratas Sprague-Dawley
3.
Brain Res ; 1835: 148918, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38588847

RESUMEN

The lateral habenula (LHb) projects to the ventral tegmental area (VTA) and dorsal raphe nuclei (DRN) that deliver dopamine (DA) and serotonin (5-HT) to cortical and limbic regions such as the medial prefrontal cortex (mPFC), hippocampus and basolateral amygdala (BLA). Dysfunctions of VTA-related mesocorticolimbic dopaminergic and DRN-related serotonergic systems contribute to non-motor symptoms in Parkinson's disease (PD). However, how the LHb affects the VTA and DRN in PD remains unclear. Here, we used electrophysiological and neurochemical approaches to explore the effects of LHb lesions on the firing activity of VTA and DRN neurons, as well as the levels of DA and 5-HT in related brain regions in unilateral 6-hydroxydopamie (6-OHDA)-induced PD rats. We found that compared to sham lesions, lesions of the LHb increased the firing rate of DA neurons in the VTA and 5-HT neurons in the DRN, but decreased the firing rate of GABAergic neurons in the same nucleus. In addition, lesions of the LHb increased the levels of DA and 5-HT in the mPFC, ventral hippocampus and BLA compared to sham lesions. These findings suggest that lesions of the LHb enhance the activity of mesocorticolimbic dopaminergic and serotonergic systems in PD.


Asunto(s)
Dopamina , Neuronas Dopaminérgicas , Núcleo Dorsal del Rafe , Habénula , Ratas Sprague-Dawley , Neuronas Serotoninérgicas , Serotonina , Área Tegmental Ventral , Animales , Área Tegmental Ventral/metabolismo , Habénula/metabolismo , Masculino , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Núcleo Dorsal del Rafe/metabolismo , Neuronas Serotoninérgicas/metabolismo , Neuronas Serotoninérgicas/fisiología , Ratas , Serotonina/metabolismo , Dopamina/metabolismo , Oxidopamina/toxicidad , Trastornos Parkinsonianos/fisiopatología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/patología , Corteza Prefrontal/metabolismo , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología
4.
Neuroscience ; 544: 88-101, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38431042

RESUMEN

Short-chain fatty acids (SCFAs) are bioactive lipids that are released into the colon as a metabolite of bacterial fermentation of dietary fibers. Beyond their function in the gastrointestinal tract, SCFAs can also have effects inthe brain, as a part of the gut-brain axis. Recent investigations into potential therapeutic interventions via the manipulation of the gut microbiome-and thus their SCFA metabolites-has been emerging as a new branch of personalized medicine,especially for mental health conditions. The current study sought to measure and localize SCFA receptors in the mouse brain. Two cell types have been implicated in the gut-brain axis: microglia and serotonergic neurons. We used fluorescentin situhybridization in brain sections from mice fed diets with different compositions of fat and fiber to quantify the mRNA levels of known gene markers of these two cell types and colocalize each with mRNA for free fatty acid receptors that bind SCFAs. We focused onmicroglia in the hippocampus and the serotonergic neurons of the dorsal raphe. We found high colocalization of SCFA receptors in both microglia and serotonergic neurons and discovered that SCFA receptor expression in the dorsal raphe is driven by fiber solubility, while SCFA receptor expression in the hippocampus is driven by fiber amount. Higher dietary fiber was associated with decreased tyrosine hydroxylase expression. Thus, our results indicate that the amount and solubility of dietary fiber can change gene expression in the brain's microglia and serotonin neurons, potentially via sensitivity to circulating levels of SCFAs produced in the gut.


Asunto(s)
Microglía , Neuronas Serotoninérgicas , Animales , Ratones , Microglía/metabolismo , Neuronas Serotoninérgicas/metabolismo , Ácidos Grasos Volátiles/metabolismo , Fibras de la Dieta/metabolismo , Encéfalo/metabolismo
5.
ACS Chem Neurosci ; 15(5): 932-943, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38377680

RESUMEN

Alzheimer's disease (AD) is a progressive degenerative disorder that results in a severe loss of brain cells and irreversible cognitive decline. Memory problems are the most recognized symptoms of AD. However, approximately 90% of patients diagnosed with AD suffer from behavioral symptoms, including mood changes and social impairment years before cognitive dysfunction. Recent evidence indicates that the dorsal raphe nucleus (DRN) is among the initial regions that show tau pathology, which is a hallmark feature of AD. The DRN harbors serotonin (5-HT) neurons, which are critically involved in mood, social, and cognitive regulation. Serotonergic impairment early in the disease process may contribute to behavioral symptoms in AD. However, the mechanisms underlying vulnerability and contribution of the 5-HT system to AD progression remain unknown. Here, we performed behavioral and electrophysiological characterizations in mice expressing a phosphorylation-prone form of human tau (hTauP301L) in 5-HT neurons. We found that pathological tau expression in 5-HT neurons induces anxiety-like behavior and alterations in stress-coping strategies in female and male mice. Female mice also exhibited social disinhibition and mild cognitive impairment in response to 5-HT neuron-specific hTauP301L expression. Behavioral alterations were accompanied by disrupted 5-HT neuron physiology in female and male hTauP301L expressing mice with exacerbated excitability disruption in females only. These data provide mechanistic insights into the brain systems and symptoms impaired early in AD progression, which is critical for disease intervention.


Asunto(s)
Neuronas , Proteínas tau , Animales , Femenino , Humanos , Masculino , Ratones , Enfermedad de Alzheimer/metabolismo , Ansiedad , Núcleo Dorsal del Rafe/metabolismo , Neuronas/metabolismo , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Proteínas tau/metabolismo
6.
Redox Biol ; 69: 103005, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38150991

RESUMEN

Major depressive disorder (MDD) is a devastating condition. Although progress has been made in the past seven decades, patients with MDD continue to receive an inadequate treatment, primarily due to the late onset of first-line antidepressant drugs and to their acute withdrawal symptoms. Resilience is the ability to rebound from adversity in a healthy manner and many people have psychological resilience. Revealing the mechanisms and identifying methods promoting resilience will hopefully lead to more effective prevention strategies and treatments for depression. In this study, we found that intermittent hypobaric hypoxia training (IHHT), a method for training pilots and mountaineers, enhanced psychological resilience in adult mice. IHHT produced a sustained antidepressant-like effect in mouse models of depression by inducing long-term (up to 3 months after this treatment) overexpression of hypoxia-inducible factor (HIF)-1α in the dorsal raphe nucleus (DRN) of adult mice. Moreover, DRN-infusion of cobalt chloride, which mimics hypoxia increasing HIF-1α expression, triggered a rapid and long-lasting antidepressant-like effect. Down-regulation of HIF-1α in the DRN serotonergic (DRN5-HT) neurons attenuated the effects of IHHT. HIF-1α translationally regulated the expression of P2X2, and conditionally knocking out P2rx2 (encodes P2X2 receptors) in DRN5-HT neurons, in turn, attenuated the sustained antidepressant-like effect of IHHT, but not its acute effect. In line with these results, a single sub-anesthetic dose of ketamine enhanced HIF-1α-P2X2 signaling, which is essential for its rapid and long-lasting antidepressant-like effect. Notably, we found that P2X2 protein levels were significantly lower in the DRN of patients with MDD than that of control subjects. Together, these findings elucidate the molecular mechanism underlying IHHT promoting psychological resilience and highlight enhancing HIF-1α-P2X2 signaling in DRN5-HT neurons as a potential avenue for screening novel therapeutic treatments for MDD.


Asunto(s)
Trastorno Depresivo Mayor , Resiliencia Psicológica , Humanos , Ratones , Animales , Núcleo Dorsal del Rafe/metabolismo , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Serotonina/farmacología , Antidepresivos/farmacología , Hipoxia , Receptores Purinérgicos P2X2/metabolismo
7.
Anal Chem ; 95(42): 15614-15620, 2023 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-37830753

RESUMEN

In brains, the serotonergic neurons are the unique resource of the neurotransmitter serotonin, which plays a pivotal role in the physiology of the brain. The dysfunction of serotonergic neurons caused by oxidative stress in the brain is closely related to the occurrence and development of various mental diseases, such as depression. As the biomarker of oxidative stress, the superoxide anion radical (O2•-) can cause oxidative damage to proteins, nucleic acids and lipids, disturbing the function of neurons and brains. A serotonin transporter (SERT) specifically expresses in serotonergic neurons, which is the biomarker of serotonergic neurons. Thus, we created two novel small molecular fluorescent probes (PA-CA and HT-CA) for imaging O2•- in serotonergic neurons of living brains of mice based on specific targeting groups of SERT. Both PA-CA and HT-CA exert excellent SERT-targetable and glorious selectivity for O2•-. Those two probes could monitor the boost of O2•- in living hsert-HEK293 cells that specifically express SERT under oxidative stress. With two-photon fluorescence imaging, we revealed for the first time that O2•- is significantly increased in serotonergic neurons in living brains of mice with depression. More importantly, proteomic analyses suggested that O2•- could oxidize cysteine and histidine in the active site of SERT, which is involved in the development of depression. This work provides new materials for living brain imaging and offers new strategy for unraveling the pathophysiology of depression.


Asunto(s)
Colorantes Fluorescentes , Neuronas Serotoninérgicas , Ratones , Humanos , Animales , Neuronas Serotoninérgicas/metabolismo , Colorantes Fluorescentes/metabolismo , Superóxidos/metabolismo , Depresión , Fluorescencia , Células HEK293 , Proteómica , Encéfalo , Biomarcadores/metabolismo
8.
Dev Neurobiol ; 83(7-8): 268-281, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37714743

RESUMEN

Serotonergic neurons produce extensively branched axons that fill most of the central nervous system, where they modulate a wide variety of behaviors. Many behavioral disorders have been correlated with defective serotonergic axon morphologies. Proper behavioral output therefore depends on the precise outgrowth and targeting of serotonergic axons during development. To direct outgrowth, serotonergic neurons utilize serotonin as a signaling molecule prior to it assuming its neurotransmitter role. This process, termed serotonin autoregulation, regulates axon outgrowth, branching, and varicosity development of serotonergic neurons. However, the receptor that mediates serotonin autoregulation is unknown. Here we asked if serotonin receptor 5-HT1A plays a role in serotonergic axon outgrowth and branching. Using cultured Drosophila serotonergic neurons, we found that exogenous serotonin reduced axon length and branching only in those expressing 5-HT1A. Pharmacological activation of 5-HT1A led to reduced axon length and branching, whereas the disruption of 5-HT1A rescued outgrowth in the presence of exogenous serotonin. Altogether this suggests that 5-HT1A is a serotonin autoreceptor in a subpopulation of serotonergic neurons and initiates signaling pathways that regulate axon outgrowth and branching during Drosophila development.


Asunto(s)
Neuronas Serotoninérgicas , Serotonina , Animales , Drosophila/metabolismo , Proyección Neuronal , Receptores de Serotonina/metabolismo , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo
9.
Nature ; 617(7962): 777-784, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37100911

RESUMEN

Associating multiple sensory cues with objects and experience is a fundamental brain process that improves object recognition and memory performance. However, neural mechanisms that bind sensory features during learning and augment memory expression are unknown. Here we demonstrate multisensory appetitive and aversive memory in Drosophila. Combining colours and odours improved memory performance, even when each sensory modality was tested alone. Temporal control of neuronal function revealed visually selective mushroom body Kenyon cells (KCs) to be required for enhancement of both visual and olfactory memory after multisensory training. Voltage imaging in head-fixed flies showed that multisensory learning binds activity between streams of modality-specific KCs so that unimodal sensory input generates a multimodal neuronal response. Binding occurs between regions of the olfactory and visual KC axons, which receive valence-relevant dopaminergic reinforcement, and is propagated downstream. Dopamine locally releases GABAergic inhibition to permit specific microcircuits within KC-spanning serotonergic neurons to function as an excitatory bridge between the previously 'modality-selective' KC streams. Cross-modal binding thereby expands the KCs representing the memory engram for each modality into those representing the other. This broadening of the engram improves memory performance after multisensory learning and permits a single sensory feature to retrieve the memory of the multimodal experience.


Asunto(s)
Encéfalo , Percepción de Color , Drosophila melanogaster , Aprendizaje , Memoria , Neuronas , Percepción Olfatoria , Animales , Encéfalo/citología , Encéfalo/fisiología , Dopamina/metabolismo , Aprendizaje/fisiología , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/fisiología , Neuronas/fisiología , Drosophila melanogaster/citología , Drosophila melanogaster/fisiología , Neuronas GABAérgicas/metabolismo , Neuronas Serotoninérgicas/metabolismo , Memoria/fisiología , Percepción Olfatoria/fisiología , Neuronas Dopaminérgicas/metabolismo , Inhibición Neural , Percepción de Color/fisiología , Odorantes/análisis
10.
Mol Metab ; 69: 101676, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36682413

RESUMEN

OBJECTIVE: Serotonin (5HT) is a well-known anorexigenic molecule, and 5HT neurons of dorsal raphe nucleus (DRN) have been implicated in suppression of feeding; however, the downstream circuitry is poorly understood. Here we explored major projections of DRN5HT neurons for their capacity to modulate feeding. METHODS: We used optogenetics to selectively activate DRN5HT axonal projections in hypothalamic and extrahypothalamic areas and monitored food intake. We next used fiber photometry to image the activity dynamics of DRN5HT axons and 5HT levels in projection areas in response feeding and metabolic hormones. Finally, we used electrophysiology to determine how DRN5HT axons affect downstream neuron activity. RESULTS: We found that selective activation of DRN5HT axons in (DRN5HT → LH) and (DRN5HT → BNST) suppresses feeding whereas activating medial hypothalamic projections has no effect. Using in vivo imaging, we found that food access and satiety hormones activate DRN5HT projections to LH where they also rapidly increase extracellular 5HT levels. Optogenetic mapping revealed that DRN5HT → LHvGAT and DRN5HT → LHvGlut2 connections are primarily inhibitory and excitatory respectively. Further, in addition to its direct action on LH neurons, we found that 5HT suppresses GABA release from presynaptic terminals arriving from AgRP neurons. CONCLUSIONS: These findings define functionally redundant forebrain circuits through which DRN5HT neurons suppress feeding and reveal that these projections can be modulated by metabolic hormones.


Asunto(s)
Núcleo Dorsal del Rafe , Neuronas Serotoninérgicas , Núcleo Dorsal del Rafe/metabolismo , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Hipotálamo/metabolismo , Hormonas
11.
Gac Med Mex ; 158(4): 182-189, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36256550

RESUMEN

INTRODUCTION: Diabetes mellitus (DM) inhibits brain serotonin biosynthesis through changes in tryptophan-5-hydroxylase (TPH) activity and expression. OBJECTIVES: To determine whether DM-induced changes in brain TPH1 or TPH2 expression and in the number of serotonergic neurons return to normal in diabetic rats treated with insulin. METHODS: Rats with streptozotocin-induced diabetes were divided in two groups: one treated with insulin and the other without treatment. On day 14, brain stems were obtained in order to quantify L-tryptophan and 5-hydroxytryptamine levels, as well as to determine TPH activity. The expression of TPH1 and TPH2 by West-ern blot, and the number of serotonergic neurons by immunohistochemistry. RESULTS: In diabetic rats, a decrease in the levels of L-tryptophan, 5-hydroxytryptamine, and TPH activity was confirmed, as well as lower TPH1 and TPH2 expression and lower numbers of serotonergic neurons. When diabetic rats were treated with insulin, L-tryptophan returned to normal, but not 5-hy-droxytryptamine, TPH expression, or the number of serotonergic neurons. CONCLUSIONS: DM chronically inhibits the synthesis of brain 5-hydroxytryptamine through changes in TPH1 and TPH2 expression and a decrease in the number of serotonergic neurons, which persist despite insulin treatment.


INTRODUCCIÓN: La diabetes mellitus (DM) inhibe la biosíntesis de serotonina cerebral mediante cambios en la actividad y expresión de la triptófano-5-hidroxilasa (TPH). OBJETIVOS: Determinar si los cambios en la expresión de TPH1 o TPH2 cerebral y en el número de neuronas serotoninérgicas causados por la DM retornan a la normalidad en las ratas con diabetes tratadas con insulina. MÉTODOS: Ratas con diabetes inducida con estreptozotocina se dividieron en dos grupos: uno tratado con insulina y otro sin tratamiento. En el día 14, se obtuvieron tallos cerebrales para cuantificar niveles de L-triptófano, 5-hidroxitriptamina y la actividad de la TPH. La expresión de TPH1 y TPH2 fue mediante Western blot y el número de neuronas serotoninérgicas por inmu­nohistoquímica. RESULTADOS: En las ratas con diabetes se confirmó disminución de los niveles de L-triptófano, 5-hidroxitriptamina y la actividad de la TPH, así como una menor expresión de TPH1 y 2 y un menor número de neuronas serotoninérgicas. Cuando las ratas diabéticas fueron tratadas con insulina, el L-triptófano regreso a la normalidad, no así la 5-hidroxitriptamina, la expresión de TPH y el número de neuronas serotoninérgicas. CONCLUSIONES: La DM inhibe crónicamente la síntesis de 5-hidroxitriptamina cerebral mediante modificaciones en la expresión de TPH1 y TPH2 y disminución de las neuronas seroto­ninérgicas, que persisten a pesar del tratamiento con insulina.


Asunto(s)
Diabetes Mellitus Experimental , Serotonina , Animales , Ratas , Serotonina/metabolismo , Triptófano/metabolismo , Núcleos del Rafe/metabolismo , Neuronas Serotoninérgicas/metabolismo , Estreptozocina/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Triptófano Hidroxilasa/metabolismo , Encéfalo/metabolismo , Insulina/metabolismo
12.
PLoS Genet ; 18(9): e1010371, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36048889

RESUMEN

The regulation of ribosome function is a conserved mechanism of growth control. While studies in single cell systems have defined how ribosomes contribute to cell growth, the mechanisms that link ribosome function to organismal growth are less clear. Here we explore this issue using Drosophila Minutes, a class of heterozygous mutants for ribosomal proteins. These animals exhibit a delay in larval development caused by decreased production of the steroid hormone ecdysone, the main regulator of larval maturation. We found that this developmental delay is not caused by decreases in either global ribosome numbers or translation rates. Instead, we show that they are due in part to loss of Rp function specifically in a subset of serotonin (5-HT) neurons that innervate the prothoracic gland to control ecdysone production. We find that these effects do not occur due to altered protein synthesis or proteostasis, but that Minute animals have reduced expression of synaptotagmin, a synaptic vesicle protein, and that the Minute developmental delay can be partially reversed by overexpression of synaptic vesicle proteins in 5-HTergic cells. These results identify a 5-HT cell-specific role for ribosomal function in the neuroendocrine control of animal growth and development.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ecdisona/metabolismo , Regulación del Desarrollo de la Expresión Génica , Larva , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Neuronas Serotoninérgicas/metabolismo , Serotonina/genética , Serotonina/metabolismo
13.
Prog Neurobiol ; 217: 102333, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35872219

RESUMEN

The neurotrophin brain-derived neurotrophic factor (BDNF) stimulates adult neurogenesis, but also influences structural plasticity and function of serotonergic neurons. Both, BDNF/TrkB signaling and the serotonergic system modulate behavioral responses to stress and can lead to pathological states when dysregulated. The two systems have been shown to mediate the therapeutic effect of antidepressant drugs and to regulate hippocampal neurogenesis. To elucidate the interplay of both systems at cellular and behavioral levels, we generated a transgenic mouse line that overexpresses BDNF in serotonergic neurons in an inducible manner. Besides displaying enhanced hippocampus-dependent contextual learning, transgenic mice were less affected by chronic social defeat stress (CSDS) compared to wild-type animals. In parallel, we observed enhanced serotonergic axonal sprouting in the dentate gyrus and increased neural stem/progenitor cell proliferation, which was uniformly distributed along the dorsoventral axis of the hippocampus. In the forced swim test, BDNF-overexpressing mice behaved similarly as wild-type mice treated with the antidepressant fluoxetine. Our data suggest that BDNF released from serotonergic projections exerts this effect partly by enhancing adult neurogenesis. Furthermore, independently of the genotype, enhanced neurogenesis positively correlated with the social interaction time after the CSDS, a measure for stress resilience.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Neuronas Serotoninérgicas , Animales , Antidepresivos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Fluoxetina/metabolismo , Fluoxetina/farmacología , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Neurogénesis/fisiología , Neuronas Serotoninérgicas/metabolismo
14.
Mol Cell Neurosci ; 121: 103750, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35697176

RESUMEN

The central serotonin2B receptor (5-HT2BR) modulates 5-HT and dopamine (DA) neuronal function in the mammalian brain and has been suggested as a potential target for the treatment of neuropsychiatric disorders involving derangements of these monoamine systems, such as schizophrenia, cocaine abuse and dependence and major depressive disorder. Studies in rats and mice yielded contrasting results on the control of 5-HT/DA networks by 5-HT2BRs, thereby leading to opposite views on the therapeutic potential of 5-HT2BR agents for treating the above disorders. These discrepancies may result from anatomo-functional differences related to a different cellular location of 5-HT2BRs in rat and mouse brain. Using immunohistochemistry, we assessed this hypothesis by examining the expression of 5-HT2BRs in 5-HT and GABAergic neurons of rats and mice within different subregions of the dorsal raphe nucleus (DRN), currently considered as the main site of action of 5-HT2B agents. Likewise, using in vivo microdialysis, we examined their functional relevance in the control of DRN 5-HT outflow, a surrogate index of 5-HT neuronal activity. In the DRN of both species, 5-HT2BRs are expressed in 5-HT cells expressing tryptophan hydroxylase 2 (TPH2), in GABAergic cells expressing glutamic acid decarboxylase 67 (GAD67), and in cells expressing both markers (GAD67 & TPH2; i.e., GABA-expressing 5-HT neurons). The proportion of 5-HT2BR-positive cells expressing only TPH2 was significantly larger in mouse than in rat DRN, whereas the opposite holds true for the expression in cells expressing GAD67 & TPH2. No major species differences were found in the dorsal and ventral subregions. In contrast, the lateral subregion exhibited large differences, with a predominant expression of 5-HT2BRs in TPH2-positive cells in mice (67.2 vs 19.9 % in rats), associated with a lower expression in GAD67 & TPH2 cells (7.9 % in mice vs 41.5 % in rats). Intra-DRN (0.1 µM) administration of the preferential 5-HT2BR agonist BW 723C86 decreased and increased DRN 5-HT outflow in rats and mice respectively, both effects being prevented by the intra-DRN perfusion of the selective 5-HT2BR antagonist RS 127445 (0.1 µM). Altogether, these results show the existence of anatomical differences in the cellular expression of 5-HT2BRs in the rat and mouse DRN, which translate into an opposite control of 5-HT outflow. Also, they highlight the relevance of the subset of GAD67-positive 5-HT neurons as a key factor responsible for the functional differences between rats and mice in terms of 5-HT neuronal activity modulation.


Asunto(s)
Núcleo Dorsal del Rafe , Receptor de Serotonina 5-HT2B , Neuronas Serotoninérgicas , Animales , Núcleo Dorsal del Rafe/metabolismo , Ratones , Ratas , Receptor de Serotonina 5-HT2B/metabolismo , Neuronas Serotoninérgicas/metabolismo , Serotonina/farmacología
15.
Neuron ; 110(14): 2268-2282.e4, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35550066

RESUMEN

Colorectal cancer stem cells (CSCs) contribute to colorectal tumorigenesis and metastasis. Colorectal CSCs reside within specialized niches and harbor self-renewal and differentiation capacities. However, the niche regulations of CSCs remain unclear. Here, we show that intestinal nerve cells are required for CSC self-renewal and colorectal tumorigenesis. Enteric serotonergic neurons produce 5-hydroxytryptamine (5-HT) to function as a modulator of CSC self-renewal. 5-HT receptors HTR1B/1D/1F are highly expressed in colorectal CSCs and engage with 5-HT to initiate Wnt/ß-catenin signaling. Mechanistically, colorectal cancer (CRC)-enriched microbiota metabolite isovalerate suppresses the enrichment of the NuRD complex onto Tph2 promoter to initiate Tph2 expression, leading to 5-HT production. 5-HT signaling is correlated with CRC severity. Blocking 5-HT signaling in mice not only inhibits the self-renewal of colorectal CSCs but also displays therapeutic efficacy against CRC tumors. Our findings reveal a cross talk between intestinal neurons and tumor cells that serves as an additional layer for CSC regulation.


Asunto(s)
Autorrenovación de las Células , Neoplasias Colorrectales , Animales , Carcinogénesis , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Ratones , Neuronas Serotoninérgicas/metabolismo , Serotonina , Vía de Señalización Wnt
16.
J Neural Transm (Vienna) ; 129(5-6): 703-711, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35420371

RESUMEN

3,4-Methylenedioxymethamphetamine (MDMA, "Ecstasy") use has been linked to persistent alterations of the brain serotonergic (5-HT) system in animal and human studies, but the molecular underpinnings are still unclear. Cytoskeletal structures such as neurofilament light chain (NfL) are promising markers of drug-induced brain toxicity and may be involved in MDMA neurotoxicity. The brain-derived neurotrophic factor (BDNF) promotes the growth and sprouting of 5-HT neurons and its differential response to MDMA administration was suggested to mediate dose- and region-dependent 5-HT damage by MDMA. However, the role of BDNF pre-treatment in preventing MDMA neurotoxicity and the potential effects of MDMA on NfL are still elusive. Therefore, a differentiated 5-HT neuronal cell line obtained from rat raphe nucleus (RN46A) was treated in vitro with either MDMA, BDNF, MDMA + BDNF, or vehicle. Cell viability (measured by MTT) and intracellular NfL levels (immunocytochemistry assay) were reduced by MDMA, but partially rescued by BDNF co-treatment. Our findings confirmed that BDNF levels can influence MDMA-induced 5-HT damage, and support BDNF to be a crucial target for neuroprotective interventions of the 5-HT system. We also provide evidence on the sensitivity of NfL to MDMA neurotoxicity, with potential implications for in-vivo monitoring of drug-induced neurotoxicity.


Asunto(s)
N-Metil-3,4-metilenodioxianfetamina , Síndromes de Neurotoxicidad , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , N-Metil-3,4-metilenodioxianfetamina/toxicidad , Ratas , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo
17.
Neuron ; 110(6): 1036-1050.e7, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35051377

RESUMEN

The nervous and endocrine systems coordinately monitor and regulate nutrient availability to maintain energy homeostasis. Sensory detection of food regulates internal nutrient availability in a manner that anticipates food intake, but sensory pathways that promote anticipatory physiological changes remain unclear. Here, we identify serotonergic (5-HT) neurons as critical mediators that transform gustatory detection by sensory neurons into the activation of insulin-producing cells and enteric neurons in Drosophila. One class of 5-HT neurons responds to gustatory detection of sugars, excites insulin-producing cells, and limits consumption, suggesting that they anticipate increased nutrient levels and prevent overconsumption. A second class of 5-HT neurons responds to gustatory detection of bitter compounds and activates enteric neurons to promote gastric motility, likely to stimulate digestion and increase circulating nutrients upon food rejection. These studies demonstrate that 5-HT neurons relay acute gustatory detection to divergent pathways for longer-term stabilization of circulating nutrients.


Asunto(s)
Proteínas de Drosophila , Gusto , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Nutrientes , Neuronas Serotoninérgicas/metabolismo , Gusto/fisiología
18.
Artículo en Inglés | MEDLINE | ID: mdl-34592387

RESUMEN

Independent from homeostatic needs, the consumption of foods originating from hyperpalatable diets is defined as hedonic eating. Hedonic eating can be observed in many forms of eating phenotypes, such as compulsive eating and stress-eating, heightening the risk of obesity development. For instance, stress can trigger the consumption of palatable foods as a type of coping strategy, which can become compulsive, particularly when developed as a habit. Although eating for pleasure is observed in multiple maladaptive eating behaviours, the current understanding of the neurobiology underlying hedonic eating remains deficient. Intriguingly, the combined orexigenic, anxiolytic and reward-seeking properties of Neuropeptide Y (NPY) ignited great interest and has positioned NPY as one of the core neuromodulators operating hedonic eating behaviours. While extensive literature exists exploring the homeostatic orexigenic and anxiolytic properties of NPY, the rewarding effects of NPY continue to be investigated. As deduced from a series of behavioural and molecular-based studies, NPY appears to motivate the consumption and enhancement of food-rewards. As a possible mechanism, NPY may modulate reward-associated monoaminergic pathways, such as the dopaminergic and serotoninergic neural networks, to modulate hedonic eating behaviours. Furthermore, potential direct and indirect NPYergic neurocircuitries connecting classical homeostatic and hedonic neuropathways may also exist involving the anti-reward centre the lateral habenula. Therefore, this review investigates the participation of NPY in orchestrating hedonic eating behaviours through the modulation of monoaminergic pathways.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Conducta Alimentaria/fisiología , Vías Nerviosas/metabolismo , Neuropéptido Y/metabolismo , Recompensa , Neuronas Serotoninérgicas/metabolismo , Adaptación Psicológica , Homeostasis , Humanos , Motivación , Obesidad/fisiopatología
19.
Behav Brain Res ; 419: 113688, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34843742

RESUMEN

Serotonin neurotransmission has been implicated in behavior deficits that occur during protracted withdrawal from opioids. In addition, studies have highlighted multiple pathways whereby serotonin (5-HT) modulates energy homeostasis, however the underlying metabolic effects of opioid withdrawal have not been investigated. A key metabolic regulator that senses the energy status of the cell and regulates fuel availability is Adenosine Monophosphate-activated Protein Kinase (AMPK). To investigate the interaction between cellular metabolism and serotonin in modulating protracted abstinence from morphine, we depleted AMPK in serotonin neurons. Morphine exposure via drinking water generates dependence in these mice, and both wildtype and serotonergic AMPK knockout mice consume similar amounts of morphine with no changes in body weight. Serotonergic AMPK contributes to baseline differences in open field and social interaction behaviors and blocks abstinence induced reductions in immobility following morphine withdrawal in the tail suspension test. Lastly, morphine locomotor sensitization is blunted in mice lacking AMPK in serotonin neurons. Taken together, our results suggest serotonergic AMPK mediates both baseline and protracted morphine withdrawal-induced behaviors.


Asunto(s)
Quinasas de la Proteína-Quinasa Activada por el AMP/metabolismo , Dependencia de Morfina/metabolismo , Dependencia de Morfina/fisiopatología , Neuronas Serotoninérgicas/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/fisiopatología , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
20.
Biochem Biophys Res Commun ; 586: 114-120, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34839189

RESUMEN

Prepulse inhibition (PPI) is a neurophysiological finding that is decreased in schizophrenia patients and has been used in pathophysiology studies of schizophrenia and the development of antipsychotic drugs. PPI is affected by several drugs including amphetamine, ketamine, and nicotinic agents, and it is reported that several brain regions and modulatory neurotransmitters are involved in PPI. Here we showed that mice with IRSp53 deletion in each dopaminergic, cholinergic, oxytocinergic, and serotoninergic modulatory neurons showed a decrease in PPI. Other than PPI, there were no other behavioral changes among IRSp53 deletion mice. Through this study, we could reconfirm that dysfunction of each modulatory neuron such as dopamine, acetylcholine, oxytocin, and serotonin can result in PPI impairment, and it should be considered that PPI could be broadly affected by changes in one of a certain kind of modulatory neurons.


Asunto(s)
Encéfalo/metabolismo , Neuronas Colinérgicas/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteínas del Tejido Nervioso/genética , Inhibición Prepulso , Neuronas Serotoninérgicas/metabolismo , Acetilcolina/metabolismo , Animales , Encéfalo/patología , Mapeo Encefálico , Neuronas Colinérgicas/patología , Dopamina/metabolismo , Neuronas Dopaminérgicas/patología , Eliminación de Gen , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Ruido , Oxitocina/metabolismo , Reflejo de Sobresalto , Neuronas Serotoninérgicas/patología , Serotonina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...