Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.840
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(12): 2574-2592.e20, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37192620

RESUMEN

Serotonin influences many aspects of animal behavior. But how serotonin acts on its diverse receptors across the brain to modulate global activity and behavior is unknown. Here, we examine how serotonin release in C. elegans alters brain-wide activity to induce foraging behaviors, like slow locomotion and increased feeding. Comprehensive genetic analyses identify three core serotonin receptors (MOD-1, SER-4, and LGC-50) that induce slow locomotion upon serotonin release and others (SER-1, SER-5, and SER-7) that interact with them to modulate this behavior. SER-4 induces behavioral responses to sudden increases in serotonin release, whereas MOD-1 induces responses to persistent release. Whole-brain imaging reveals widespread serotonin-associated brain dynamics, spanning many behavioral networks. We map all sites of serotonin receptor expression in the connectome, which, together with synaptic connectivity, helps predict which neurons show serotonin-associated activity. These results reveal how serotonin acts at defined sites across a connectome to modulate brain-wide activity and behavior.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Caenorhabditis elegans/metabolismo , Serotonina/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Conducta Animal/fisiología , Encéfalo/metabolismo
2.
Cell ; 171(7): 1649-1662.e10, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29198526

RESUMEN

Animals generate complex patterns of behavior across development that may be shared or unique to individuals. Here, we examine the contributions of developmental programs and individual variation to behavior by monitoring single Caenorhabditis elegans nematodes over their complete developmental trajectories and quantifying their behavior at high spatiotemporal resolution. These measurements reveal reproducible trajectories of spontaneous foraging behaviors that are stereotyped within and between developmental stages. Dopamine, serotonin, the neuropeptide receptor NPR-1, and the TGF-ß peptide DAF-7 each have stage-specific effects on behavioral trajectories, implying the existence of a modular temporal program controlled by neuromodulators. In addition, a fraction of individuals within isogenic populations raised in controlled environments have consistent, non-genetic behavioral biases that persist across development. Several neuromodulatory systems increase or decrease the degree of non-genetic individuality to shape sustained patterns of behavior across the population.


Asunto(s)
Variación Biológica Individual , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/fisiología , Neuropéptidos/metabolismo , Animales , Conducta Animal , Dopamina/metabolismo , Regulación de la Expresión Génica , Larva/fisiología , Neuroimagen/instrumentación , Neuroimagen/métodos , Neuropéptidos/genética , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo
3.
Cell ; 163(3): 533, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26496594

RESUMEN

Flibanserin acts at cortical, limbic, hypothalamic, and brainstem nuclei to inhibit serotonin release by binding to 5-HT1A autoreceptors and block postsynaptic action of serotonin at 5-HT2A receptors. This gradually disinhibits the turnover of other monoamines like dopamine and noradrenaline that are critical for sexual desire.


Asunto(s)
Bencimidazoles/uso terapéutico , Antagonistas de la Serotonina/uso terapéutico , Disfunciones Sexuales Psicológicas/tratamiento farmacológico , Encéfalo/metabolismo , Humanos , Receptores de Serotonina/metabolismo , Disfunciones Sexuales Psicológicas/metabolismo
4.
Mol Cell ; 82(14): 2681-2695.e6, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35714614

RESUMEN

Serotonin (or 5-hydroxytryptamine, 5-HT) is an important neurotransmitter that activates 12 different G protein-coupled receptors (GPCRs) through selective coupling of Gs, Gi, or Gq proteins. The structural basis for G protein subtype selectivity by these GPCRs remains elusive. Here, we report the structures of the serotonin receptors 5-HT4, 5-HT6, and 5-HT7 with Gs, and 5-HT4 with Gi1. The structures reveal that transmembrane helices TM5 and TM6 alternate lengths as a macro-switch to determine receptor's selectivity for Gs and Gi, respectively. We find that the macro-switch by the TM5-TM6 length is shared by class A GPCR-G protein structures. Furthermore, we discover specific residues within TM5 and TM6 that function as micro-switches to form specific interactions with Gs or Gi. Together, these results present a common mechanism of Gs versus Gi protein coupling selectivity or promiscuity by class A GPCRs and extend the basis of ligand recognition at serotonin receptors.


Asunto(s)
Receptores Acoplados a Proteínas G , Serotonina , Proteínas de Unión al GTP/metabolismo , Ligandos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo
5.
Proc Natl Acad Sci U S A ; 120(14): e2209917120, 2023 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-36989299

RESUMEN

While most therapeutic research on G-protein-coupled receptors (GPCRs) focuses on receptor activation by (endogenous) agonists, significant therapeutic potential exists through agonist-independent intrinsic constitutive activity that can occur in various physiological and pathophysiological settings. For example, inhibiting the constitutive activity of 5-HT6R-a receptor that is found almost exclusively in the brain and mediates excitatory neurotransmission-has demonstrated a therapeutic effect on cognitive/memory impairment associated with several neuropsychiatric disorders. However, the structural basis of such constitutive activity remains unclear. Here, we present a cryo-EM structure of serotonin-bound human 5-HT6R-Gs heterotrimer at 3.0-Å resolution. Detailed analyses of the structure complemented by comprehensive interrogation of signaling illuminate key structural determinants essential for constitutive 5-HT6R activity. Additional structure-guided mutagenesis leads to a nanobody mimic Gαs for 5-HT6R that can reduce its constitutive activity. Given the importance of 5-HT6R for a large number of neuropsychiatric disorders, insights derived from these studies will accelerate the design of more effective medications, and shed light on the molecular basis of constitutive activity.


Asunto(s)
Receptores de Serotonina , Serotonina , Humanos , Receptores de Serotonina/metabolismo , Encéfalo/metabolismo , Transducción de Señal
6.
Genes Dev ; 32(3-4): 258-270, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29491136

RESUMEN

Tissue-tissue communications are integral to organismal aging, orchestrating a body-wide aging process. The brain plays a key role in this process by detecting and processing signals from the environment and then communicating them to distal tissues such as the gut to regulate longevity. How this is achieved, however, is poorly understood. Here, using Caenorhabditis elegans as a model, we identified two distinct neuroendocrine signaling circuits by which the worm nervous system senses cool and warm environmental temperatures through cool- and warm-sensitive neurons and then signals the gut to extend and shorten life span, respectively. The prolongevity "cool" circuit uses the small neurotransmitters glutamate and serotonin, whereas the anti-longevity "warm" circuit is mediated by insulin-like neuropeptides. Both types of neuroendocrine signals converge on the gut through their cognate receptors to differentially regulate the transcription factor DAF-16/FOXO, leading to opposing outcomes in longevity. Our study illustrates how the brain detects and processes environmental signals to bidirectionally regulate longevity by signaling the gut.


Asunto(s)
Encéfalo/fisiología , Mucosa Intestinal/metabolismo , Longevidad/fisiología , Neuronas/fisiología , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Factores de Transcripción Forkhead/metabolismo , Ácido Glutámico/metabolismo , Neuropéptidos/metabolismo , Receptor de Insulina/metabolismo , Receptores de Glutamato/fisiología , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Transducción de Señal , Transmisión Sináptica , Temperatura
7.
Mol Psychiatry ; 29(3): 671-685, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38177350

RESUMEN

Acquired brain injury (ABI), such as traumatic brain injury and stroke, is a leading cause of disability worldwide, resulting in debilitating acute and chronic symptoms, as well as an increased risk of developing neurological and neurodegenerative disorders. These symptoms can stem from various neurophysiological insults, including neuroinflammation, oxidative stress, imbalances in neurotransmission, and impaired neuroplasticity. Despite advancements in medical technology and treatment interventions, managing ABI remains a significant challenge. Emerging evidence suggests that psychedelics may rapidly improve neurobehavioral outcomes in patients with various disorders that share physiological similarities with ABI. However, research specifically focussed on psychedelics for ABI is limited. This narrative literature review explores the neurochemical properties of psychedelics as a therapeutic intervention for ABI, with a focus on serotonin receptors, sigma-1 receptors, and neurotrophic signalling associated with neuroprotection, neuroplasticity, and neuroinflammation. The promotion of neuronal growth, cell survival, and anti-inflammatory properties exhibited by psychedelics strongly supports their potential benefit in managing ABI. Further research and translational efforts are required to elucidate their therapeutic mechanisms of action and to evaluate their effectiveness in treating the acute and chronic phases of ABI.


Asunto(s)
Lesiones Encefálicas , Alucinógenos , Plasticidad Neuronal , Humanos , Alucinógenos/farmacología , Alucinógenos/uso terapéutico , Plasticidad Neuronal/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Animales , Receptores de Serotonina/metabolismo , Receptores de Serotonina/efectos de los fármacos , Receptores sigma/metabolismo , Receptor Sigma-1 , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
8.
Nature ; 565(7738): 246-250, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30602786

RESUMEN

In addition to maintaining immune tolerance, FOXP3+ regulatory T (Treg) cells perform specialized functions in tissue homeostasis and remodelling1,2. However, the characteristics and functions of brain Treg cells are not well understood because there is a low number of Treg cells in the brain under normal conditions. Here we show that there is massive accumulation of Treg cells in the mouse brain after ischaemic stroke, and this potentiates neurological recovery during the chronic phase of ischaemic brain injury. Although brain Treg cells are similar to Treg cells in other tissues such as visceral adipose tissue and muscle3-5, they are apparently distinct and express unique genes related to the nervous system including Htr7, which encodes the serotonin receptor 5-HT7. The amplification of brain Treg cells is dependent on interleukin (IL)-2, IL-33, serotonin and T cell receptor recognition, and infiltration into the brain is driven by the chemokines CCL1 and CCL20. Brain Treg cells suppress neurotoxic astrogliosis by producing amphiregulin, a low-affinity epidermal growth factor receptor (EGFR) ligand. Stroke is a leading cause of neurological disability, and there are currently few effective recovery methods other than rehabilitation during the chronic phase. Our findings suggest that Treg cells and their products may provide therapeutic opportunities for neuronal protection against stroke and neuroinflammatory diseases.


Asunto(s)
Astrocitos/patología , Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Gliosis/patología , Neuroprotección/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Animales , Encéfalo/citología , Encéfalo/inmunología , Movimiento Celular , Proliferación Celular , Quimiocina CCL1/inmunología , Quimiocina CCL20/inmunología , Interleucina-2/inmunología , Interleucina-33/inmunología , Interleucina-6/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/inmunología , Receptores CCR/metabolismo , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Factor de Transcripción STAT3/metabolismo , Serotonina/metabolismo , Transducción de Señal , Linfocitos T Reguladores/metabolismo
9.
Cereb Cortex ; 34(6)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38904079

RESUMEN

Serotonin (5-HT) regulates working memory within the prefrontal cortex network, which is crucial for understanding obsessive-compulsive disorder. However, the mechanisms how network dynamics and serotonin interact in obsessive-compulsive disorder remain elusive. Here, we incorporate 5-HT receptors (5-HT1A, 5-HT2A) and dopamine receptors into a multistable prefrontal cortex network model, replicating the experimentally observed inverted U-curve phenomenon. We show how the two 5-HT receptors antagonize neuronal activity and modulate network multistability. Reduced binding of 5-HT1A receptors increases global firing, while reduced binding of 5-HT2A receptors deepens attractors. The obtained results suggest reward-dependent synaptic plasticity mechanisms may attenuate 5-HT related network impairments. Integrating serotonin-mediated dopamine release into circuit, we observe that decreased serotonin concentration triggers the network into a deep attractor state, expanding the domain of attraction of stable nodes with high firing rate, potentially causing aberrant reverse learning. This suggests a hypothesis wherein elevated dopamine concentrations in obsessive-compulsive disorder might result from primary deficits in serotonin levels. Findings of this work underscore the pivotal role of serotonergic dysregulation in modulating synaptic plasticity through dopamine pathways, potentially contributing to learned obsessions. Interestingly, serotonin reuptake inhibitors and antidopaminergic potentiators can counteract the over-stable state of high-firing stable points, providing new insights into obsessive-compulsive disorder treatment.


Asunto(s)
Trastorno Obsesivo Compulsivo , Corteza Prefrontal , Serotonina , Corteza Prefrontal/metabolismo , Trastorno Obsesivo Compulsivo/fisiopatología , Trastorno Obsesivo Compulsivo/metabolismo , Serotonina/metabolismo , Humanos , Dopamina/metabolismo , Modelos Neurológicos , Receptores Dopaminérgicos/metabolismo , Red Nerviosa/metabolismo , Red Nerviosa/fisiopatología , Simulación por Computador , Receptor de Serotonina 5-HT2A/metabolismo , Receptores de Serotonina/metabolismo , Plasticidad Neuronal/fisiología , Receptor de Serotonina 5-HT1A/metabolismo
10.
Proc Natl Acad Sci U S A ; 119(11): e2115533119, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35254908

RESUMEN

SignificancePhysiological stress triggers avoidance behavior, allowing the animals to stay away from potential threats and optimize their chance of survival. Mitochondrial disruption, a common physiological stress in diverse species, induces the nematode Caenorhabditis elegans to avoid non-pathogenic bacteria through a serotonergic neuronal circuit. We find that distinct neurons, communicated through serotonin and a specific serotonin receptor, are required for the formation and retrieval of this learned aversive behavior. This learned avoidance behavior is associated with increased serotonin synthesis, altered neuronal response property, and reprogramming of locomotion patterns. The circuit and neuromodulatory mechanisms described here offer important insights for stress-induced avoidance behavior.


Asunto(s)
Caenorhabditis elegans/fisiología , Mitocondrias/metabolismo , Receptores de Serotonina/metabolismo , Neuronas Serotoninérgicas/fisiología , Serotonina/metabolismo , Estrés Fisiológico , Animales , Reacción de Prevención , Interacciones Huésped-Patógeno , Interneuronas/metabolismo , Aprendizaje
11.
J Biol Chem ; 299(9): 105176, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37599003

RESUMEN

Substance abuse is on the rise, and while many people may use illicit drugs mainly due to their rewarding effects, their societal impact can range from severe, as is the case for opioids, to promising, as is the case for psychedelics. Common with all these drugs' mechanisms of action are G protein-coupled receptors (GPCRs), which lie at the center of how these drugs mediate inebriation, lethality, and therapeutic effects. Opioids like fentanyl, cannabinoids like tetrahydrocannabinol, and psychedelics like lysergic acid diethylamide all directly bind to GPCRs to initiate signaling which elicits their physiological actions. We herein review recent structural studies and provide insights into the molecular mechanisms of opioids, cannabinoids, and psychedelics at their respective GPCR subtypes. We further discuss how such mechanistic insights facilitate drug discovery, either toward the development of novel therapies to combat drug abuse or toward harnessing therapeutic potential.


Asunto(s)
Drogas Ilícitas , Receptores Acoplados a Proteínas G , Humanos , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacología , Cannabinoides/metabolismo , Cannabinoides/farmacología , Alucinógenos/metabolismo , Alucinógenos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Drogas Ilícitas/metabolismo , Drogas Ilícitas/farmacología , Modelos Moleculares , Receptores de Serotonina/metabolismo , Desarrollo de Medicamentos/normas
12.
Physiol Rev ; 97(2): 553-622, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28179394

RESUMEN

Plaguing humans for more than two millennia, manifest on every continent studied, and with more than one billion patients having an attack in any year, migraine stands as the sixth most common cause of disability on the planet. The pathophysiology of migraine has emerged from a historical consideration of the "humors" through mid-20th century distraction of the now defunct Vascular Theory to a clear place as a neurological disorder. It could be said there are three questions: why, how, and when? Why: migraine is largely accepted to be an inherited tendency for the brain to lose control of its inputs. How: the now classical trigeminal durovascular afferent pathway has been explored in laboratory and clinic; interrogated with immunohistochemistry to functional brain imaging to offer a roadmap of the attack. When: migraine attacks emerge due to a disorder of brain sensory processing that itself likely cycles, influenced by genetics and the environment. In the first, premonitory, phase that precedes headache, brain stem and diencephalic systems modulating afferent signals, light-photophobia or sound-phonophobia, begin to dysfunction and eventually to evolve to the pain phase and with time the resolution or postdromal phase. Understanding the biology of migraine through careful bench-based research has led to major classes of therapeutics being identified: triptans, serotonin 5-HT1B/1D receptor agonists; gepants, calcitonin gene-related peptide (CGRP) receptor antagonists; ditans, 5-HT1F receptor agonists, CGRP mechanisms monoclonal antibodies; and glurants, mGlu5 modulators; with the promise of more to come. Investment in understanding migraine has been very successful and leaves us at a new dawn, able to transform its impact on a global scale, as well as understand fundamental aspects of human biology.


Asunto(s)
Encéfalo/metabolismo , Péptido Relacionado con Gen de Calcitonina/metabolismo , Cognición/fisiología , Trastornos Migrañosos/fisiopatología , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Animales , Humanos , Trastornos Migrañosos/diagnóstico , Trastornos Migrañosos/metabolismo , Receptores de Serotonina/metabolismo , Receptor de Serotonina 5-HT1F
13.
J Neurochem ; 168(6): 1097-1112, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38323657

RESUMEN

Microdosing ketamine is a novel antidepressant for treatment-resistant depression. Traditional antidepressants, like selective serotonin reuptake inhibitors (SSRIs), inhibit serotonin reuptake, but it is not clear if ketamine shows a similar mechanism. Here, we tested the effects of feeding ketamine and SSRIs to Drosophila melanogaster larvae, which has a similar serotonin system to mammals and is a good model to track depressive behaviors, such as locomotion and feeding. Fast-scan cyclic voltammetry (FSCV) was used to measure optogenetically stimulated serotonin changes, and locomotion tracking software and blue dye feeding to monitor behavior. We fed larvae various doses (1-100 mM) of antidepressants for 24 h and found that 1 mM ketamine did not affect serotonin, but increased locomotion and feeding. Low doses (≤10 mM) of escitalopram and fluoxetine inhibited dSERT and also increased feeding and locomotion behaviors. At 100 mM, ketamine inhibited dSERT and increased serotonin concentrations, but decreased locomotion and feeding because of its anesthetic properties. Since microdosing ketamine causes behavioral effects, we further investigated behavioral changes with a SERT16 mutant and low doses of other NMDA receptor antagonists and 5-HT1A and 2 agonists. Feeding and locomotion changes were similar to ketamine in the mutant, and we found NMDA receptor antagonism increased feeding, while serotonin receptor agonism increased locomotion, which could explain these effects with ketamine. Ultimately, this work shows that Drosophila is a good model to discern antidepressant mechanisms, and that ketamine does not work on dSERT like SSRIs, but effects behavior with other mechanisms that should be investigated further.


Asunto(s)
Drosophila melanogaster , Ketamina , Locomoción , Receptores de Serotonina , Inhibidores Selectivos de la Recaptación de Serotonina , Animales , Ketamina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Receptores de Serotonina/metabolismo , Receptores de Serotonina/efectos de los fármacos , Locomoción/efectos de los fármacos , Receptores de Glutamato/metabolismo , Receptores de Glutamato/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Serotonina/metabolismo , Conducta Alimentaria/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Larva , Fluoxetina/farmacología , Antidepresivos/farmacología
14.
Am J Physiol Gastrointest Liver Physiol ; 327(1): G57-G69, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38713616

RESUMEN

Inflammatory bowel disease (IBD) encompasses several debilitating chronic gastrointestinal (GI) inflammatory disorders, including Crohn's disease and ulcerative colitis. In both conditions, mucosal inflammation is a key clinical presentation associated with altered serotonin (5-hydroxytryptamine or 5-HT) signaling. This altered 5-HT signaling is also found across various animal models of colitis. Of the 14 known receptor subtypes, 5-HT receptor type 7 (5-HT7) is one of the most recently discovered. We previously reported that blocking 5-HT signaling with either a selective 5-HT7 receptor antagonist (SB-269970) or genetic ablation alleviated intestinal inflammation in murine experimental models of colitis. Here, we developed novel antagonists, namely, MC-170073 and MC-230078, which target 5-HT7 receptors with high selectivity. We also investigated the in vivo efficacy of these antagonists in experimental colitis by using dextran sulfate sodium (DSS) and the transfer of CD4+CD45RBhigh T cells to induce intestinal inflammation. Inhibition of 5-HT7 receptor signaling with the antagonists, MC-170073 and MC-230078, ameliorated intestinal inflammation in both acute and chronic colitis models, which was accompanied by lower histopathological damage and diminished levels of proinflammatory cytokines compared with vehicle-treated controls. Together, the data reveal that the pharmacological inhibition of 5-HT7 receptors by these selective antagonists ameliorates the severity of colitis across various experimental models and may, in the future, serve as a potential treatment option for patients with IBD. In addition, these findings support that 5-HT7 is a viable therapeutic target for IBD.NEW & NOTEWORTHY This study demonstrates that the novel highly selective 5-HT7 receptor antagonists, MC-170073 and MC-230078, significantly alleviated the severity of colitis across models of experimental colitis. These findings suggest that inhibition of 5-HT7 receptor signaling by these new antagonists may serve as an alternative mode of treatment to diminish symptomology in those with inflammatory bowel disease.


Asunto(s)
Colitis , Receptores de Serotonina , Antagonistas de la Serotonina , Animales , Receptores de Serotonina/metabolismo , Receptores de Serotonina/efectos de los fármacos , Colitis/tratamiento farmacológico , Colitis/inmunología , Colitis/patología , Ratones , Antagonistas de la Serotonina/farmacología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Sulfato de Dextran , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/inmunología , Transducción de Señal/efectos de los fármacos , Índice de Severidad de la Enfermedad , Colon/efectos de los fármacos , Colon/patología , Colon/metabolismo , Colon/inmunología , Masculino
15.
Cell Commun Signal ; 22(1): 233, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38641599

RESUMEN

BACKGROUND: Multiple neurodegenerative diseases are induced by the formation and deposition of protein aggregates. In particular, the microtubule-associated protein Tau leads to the development of so-called tauopathies characterized by the aggregation of hyperphosphorylated Tau within neurons. We recently showed that the constitutive activity of the serotonin receptor 7 (5-HT7R) is required for Tau hyperphosphorylation and aggregation through activation of the cyclin-dependent kinase 5 (CDK5). We also demonstrated physical interaction between 5-HT7R and CDK5 at the plasma membrane suggesting that the 5-HT7R/CDK5 complex is an integral part of the signaling network involved in Tau-mediated pathology. METHODS: Using biochemical, microscopic, molecular biological, computational and AI-based approaches, we investigated structural requirements for the formation of 5-HT7R/CDK5 complex. RESULTS: We demonstrated that 5-HT7R domains responsible for coupling to Gs proteins are not involved in receptor interaction with CDK5. We also created a structural model of the 5-HT7R/CDK5 complex and refined the interaction interface. The model predicted two conserved phenylalanine residues, F278 and F281, within the third intracellular loop of 5-HT7R to be potentially important for complex formation. While site-directed mutagenesis of these residues did not influence Gs protein-mediated receptor signaling, replacement of both phenylalanines by alanine residues significantly reduced 5-HT7R/CDK5 interaction and receptor-mediated CDK5 activation, leading to reduced Tau hyperphosphorylation and aggregation. Molecular dynamics simulations of 5-HT7R/CDK5 complex for wild-type and receptor mutants confirmed binding interface stability of the initial model. CONCLUSIONS: Our results provide a structural basis for the development of novel drugs targeting the 5-HT7R/CDK5 interaction interface for the selective treatment of Tau-related disorders, including frontotemporal dementia and Alzheimer's disease.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina , Activación Enzimática , Receptores de Serotonina , Humanos , Enfermedad de Alzheimer/metabolismo , Quinasa 5 Dependiente de la Ciclina/química , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Fosforilación , Receptores de Serotonina/química , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Transducción de Señal
16.
Pharmacol Res ; 203: 107154, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38521286

RESUMEN

Serotonin, while conventionally recognized as a neurotransmitter in the CNS, has recently gained attention for its role in the kidney. Specifically, serotonin is not only synthesized in the kidney, but it also regulates glomerular function, vascular resistance, and mitochondrial homeostasis. Because of serotonin's importance to mitochondrial health, this review is focused on the role of serotonin and its receptors in mitochondrial function in the context of acute kidney injury, chronic kidney disease, and diabetic kidney disease, all of which are characterized by mitochondrial dysfunction and none of which has approved pharmacological treatments. Evidence indicates that activation of certain serotonin receptors can stimulate mitochondrial biogenesis (MB) and restore mitochondrial homeostasis, resulting in improved renal function. Serotonin receptor agonists that induce MB are therefore of interest as potential therapeutic strategies for renal injury and disease. SIGNIFICANCE STATEMENT: Mitochondrial dysfunction is associated with many human renal diseases such as acute kidney injury, chronic kidney disease, and diabetic kidney disease, which are associated with increased morbidity and mortality. Unfortunately, none of these pathologies has an FDA-approved pharmacological intervention, underscoring the urgency of identifying new therapeutics for such disorders. Studies show that induction of mitochondrial biogenesis via serotonin (5-hydroxytryptamine, 5-HT) receptors reduces kidney injury markers, restores mitochondrial and renal function after kidney injury, and decreases mortality, suggesting that targeting 5-HT receptors may be a promising therapeutic avenue for mitochondrial dysfunction in kidney diseases. While numerous reviews describe the importance of mitochondria and mitochondrial quality control mechanisms in kidney disease, the relevance of 5-HT receptor-mediated mitochondrial metabolic modulation in the kidney has yet to be thoroughly explored.


Asunto(s)
Enfermedades Renales , Mitocondrias , Serotonina , Animales , Humanos , Riñón/metabolismo , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Biogénesis de Organelos , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Agonistas de Receptores de Serotonina/farmacología , Agonistas de Receptores de Serotonina/uso terapéutico
17.
Nature ; 561(7721): 117-121, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30150774

RESUMEN

Differences between female and male brains exist across the animal kingdom and extend from molecular to anatomical features. Here we show that sexually dimorphic anatomy, gene expression and function in the nervous system can be modulated by past experiences. In the nematode Caenorhabditis elegans, sexual differentiation entails the sex-specific pruning of synaptic connections between neurons that are shared by both sexes, giving rise to sexually dimorphic circuits in adult animals1. We discovered that starvation during juvenile stages is memorized in males to suppress the emergence of sexually dimorphic synaptic connectivity. These circuit changes result in increased chemosensory responsiveness in adult males following juvenile starvation. We find that an octopamine-mediated starvation signal dampens the production of serotonin (5-HT) to convey the memory of starvation. Serotonin production is monitored by a 5-HT1A serotonin receptor homologue that acts cell-autonomously to promote the pruning of sexually dimorphic synaptic connectivity under well-fed conditions. Our studies demonstrate how life history shapes neurotransmitter production, synaptic connectivity and behavioural output in a sexually dimorphic circuit.


Asunto(s)
Caenorhabditis elegans/citología , Caenorhabditis elegans/fisiología , Privación de Alimentos/fisiología , Plasticidad Neuronal , Neuronas/metabolismo , Serotonina/metabolismo , Caracteres Sexuales , Transducción de Señal , Envejecimiento/fisiología , Animales , Conducta Animal , Proteínas de Caenorhabditis elegans/metabolismo , Ingestión de Alimentos/fisiología , Femenino , Masculino , Octopamina/metabolismo , Receptor de Serotonina 5-HT1A/metabolismo , Receptores de Serotonina/metabolismo , Serotonina/biosíntesis , Factores de Tiempo
18.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33495318

RESUMEN

Clinical studies have reported that the psychedelic lysergic acid diethylamide (LSD) enhances empathy and social behavior (SB) in humans, but its mechanism of action remains elusive. Using a multidisciplinary approach including in vivo electrophysiology, optogenetics, behavioral paradigms, and molecular biology, the effects of LSD on SB and glutamatergic neurotransmission in the medial prefrontal cortex (mPFC) were studied in male mice. Acute LSD (30 µg/kg) injection failed to increase SB. However, repeated LSD (30 µg/kg, once a day, for 7 days) administration promotes SB, without eliciting antidepressant/anxiolytic-like effects. Optogenetic inhibition of mPFC excitatory neurons dramatically inhibits social interaction and nullifies the prosocial effect of LSD. LSD potentiates the α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) and 5-HT2A, but not N-methyl-D-aspartate (NMDA) and 5-HT1A, synaptic responses in the mPFC and increases the phosphorylation of the serine-threonine protein kinases Akt and mTOR. In conditional knockout mice lacking Raptor (one of the structural components of the mTORC1 complex) in excitatory glutamatergic neurons (Raptorf/f:Camk2alpha-Cre), the prosocial effects of LSD and the potentiation of 5-HT2A/AMPA synaptic responses were nullified, demonstrating that LSD requires the integrity of mTORC1 in excitatory neurons to promote SB. Conversely, in knockout mice lacking Raptor in GABAergic neurons of the mPFC (Raptorf/f:Gad2-Cre), LSD promotes SB. These results indicate that LSD selectively enhances SB by potentiating mPFC excitatory transmission through 5-HT2A/AMPA receptors and mTOR signaling. The activation of 5-HT2A/AMPA/mTORC1 in the mPFC by psychedelic drugs should be explored for the treatment of mental diseases with SB impairments such as autism spectrum disorder and social anxiety disorder.


Asunto(s)
Conducta Animal/efectos de los fármacos , Dietilamida del Ácido Lisérgico/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Conducta Social , Transmisión Sináptica/efectos de los fármacos , Animales , Reacción de Prevención/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Optogenética , Fosforilación/efectos de los fármacos , Corteza Prefrontal/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Receptores AMPA/agonistas , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Serotonina/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
19.
Biochem Biophys Res Commun ; 671: 327-334, 2023 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-37327704

RESUMEN

The serotonin receptor 5-HT6R is an important G-protein-coupled receptor (GPCR) that involved in essential functions within the central and peripheral nervous systems and is linked to various psychiatric disorders. Selective activation of 5-HT6R promotes neural stem cell regeneration activity. As a 5-HT6R selective agonist, 2-(5 chloro-2-methyl-1H-indol-3-yl)-N, N-dimethylethanolamine (ST1936) has been widely used to investigate the functions of the 5-HT6R. The molecular mechanism of how ST1936 is recognized by 5-HT6R and how it effectively couples with Gs remain unclear. Here, we reconstituted the ST1936-5-HT6R-Gs complex in vitro and solved its cryo-electron microscopy structure at 3.1 Å resolution. Further structural analysis and mutational studies facilitated us to identify the residues of the Y3107.43 and "toggle switch" W2816.48 of the 5-HT6R contributed to the higher efficacy of ST1936 compared with 5-HT. By uncovering the structural foundation of how 5-HT6R specifically recognizes agonists and elucidating the molecular process of G protein activation, our discoveries offer valuable insights and pave the way for the development of promising 5-HT6R agonists.


Asunto(s)
Receptores de Serotonina , Serotonina , Humanos , Microscopía por Crioelectrón , Receptores de Serotonina/metabolismo , Indoles
20.
Microvasc Res ; 148: 104516, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36889668

RESUMEN

Control of microvascular reactivity by 5-hydroxytryptamine (5-HT; serotonin) is complex and may depend on vascular bed type and 5-HT receptors. 5-HT receptors consist of seven families (5-HT1-5-HT7), with 5-HT2 predominantly mediating renal vasoconstriction. Cyclooxygenase (COX) and smooth muscle intracellular Ca2+ levels ([Ca2+]i) have been implicated in 5-HT-induced vascular reactivity. Although 5-HT receptor expression and circulating 5-HT levels are known to be dependent on postnatal age, control of neonatal renal microvascular function by 5-HT is unclear. In the present study, we demonstrate that 5-HT stimulated human TRPV4 transiently expressed in Chinese hamster ovary cells. 5-HT2A is the predominant 5-HT2 receptor subtype in freshly isolated neonatal pig renal microvascular smooth muscle cells (SMCs). HC-067047 (HC), a selective TRPV4 blocker, attenuated cation currents induced by 5-HT in the SMCs. HC also inhibited the 5-HT-induced increase in renal microvascular [Ca2+]i and constriction. Intrarenal artery infusion of 5-HT had minimal effects on systemic hemodynamics but reduced renal blood flow (RBF) and increased renal vascular resistance (RVR) in the pigs. Transdermal measurement of glomerular filtration rate (GFR) indicated that kidney infusion of 5-HT reduced GFR. HC and 5-HT2 receptor antagonist ritanserin attenuated 5-HT effects on RBF, RVR, and GFR. Moreover, the serum and urinary COX-1 and COX-2 levels in 5-HT-treated piglets were unchanged compared with the control. These data suggest that activation of renal microvascular SMC TRPV4 channels by 5-HT impairs kidney function in neonatal pigs independently of COX production.


Asunto(s)
Músculo Liso Vascular , Serotonina , Recién Nacido , Cricetinae , Animales , Humanos , Porcinos , Músculo Liso Vascular/metabolismo , Canales Catiónicos TRPV/metabolismo , Células CHO , Cricetulus , Riñón/irrigación sanguínea , Receptores de Serotonina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA