Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Eur J Anaesthesiol ; 38(Suppl 2): S97-S105, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34170884

RESUMEN

BACKGROUND: Serratus anterior plane blocks (SAPBs) and thoracic paravertebral blocks (TPVBs) can both be used for video-assisted thoracic surgery. However, it remains unknown whether the analgesic efficacy of a SAPB is comparable to that of a TPVB. OBJECTIVE: We tested the primary hypothesis that SAPBs provide noninferior analgesia compared with TPVBs for video-assisted thoracic surgery. DESIGN: A noninferiority randomised trial. SETTING: Shanghai Chest Hospital, between August 2018 and November 2018. PATIENTS: Ninety patients scheduled for video-assisted thoracic lobectomy or segmentectomy were randomised. Patients were excluded if they were unable to perform the visual analogue pain scale, or surgery was converted to thoracotomy. INTERVENTIONS: Blocks were performed after induction of general anaesthesia. The three groups were paravertebral blocks (n = 30); serratus anterior plane blocks (n = 29); and general anaesthesia alone (n = 30). PRIMARY OUTCOME MEASURES: Visual analogue pain scores (0 to 10 cm) at rest and while coughing, and Prince-Henry pain scores (0 to 4 points) were used to assess postoperative analgesia at 2, 24 and 48 h after surgery. We assessed the noninferiority of SAPBs with TPVBs on all three primary pain outcomes using a delta of 1 cm or one point as appropriate. RESULTS: The mean difference (95% confidence intervals) in visual analogue scores between the SAPBs and TPVBs was -0.04 (-0.10 to 0.03) cm at rest, -0.22 (-0.43 to -0.01) cm during coughing and -0.10 (-0.25 to 0.05) for Prince-Henry pain scores. As the upper limit of the confidence intervals were less than 1 (all P < 0.001), noninferiority was claimed for all three primary outcomes. Compared with general anaesthesia alone, the VAS scores at rest and while coughing, and the Prince-Henry pain scores for the two blocks were significantly lower during the initial 2 h after surgery. CONCLUSIONS: Serratus anterior plane blocks are quicker and easier to perform than paravertebral blocks and provide comparable analgesia in patients having video-assisted thoracic surgery. Both blocks provided analgesia that was superior to general anaesthesia alone during the initial 2 h after surgery. TRIAL REGISTRATION: Chinese Clinical Trial Registry, identifier: ChiCTR1800017671.


Asunto(s)
Dolor Agudo , Bloqueo Nervioso , China/epidemiología , Humanos , Dolor Postoperatorio/diagnóstico , Dolor Postoperatorio/etiología , Dolor Postoperatorio/prevención & control , Cirugía Torácica Asistida por Video
2.
Eur J Anaesthesiol ; 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32205572

RESUMEN

BACKGROUND: Serratus anterior and paravertebral blocks can both be used for video-assisted thoracic surgery. However, serratus anterior blocks are easier to perform, and possibly safer. We therefore tested the primary hypothesis that serratus anterior plane blocks and thoracic paravertebral blocks provide comparable analgesia for video-assisted thoracic surgery. Secondarily, we tested the hypothesis that both blocks lengthen the time to onset of surgical pain and reduce the need for rescue tramadol. METHODS: Patients having video-assisted thoracic lobectomy or segmentectomy were randomly allocated to ultrasound-guided thoracic paravertebral blocks, n = 30; ultrasound-guided serratus anterior plane blocks, n = 30; or, general anaesthesia alone, n = 30. Visual analogue pain scores analogue pain scores at rest, during coughing and Prince-Henry pain scores were used to assess postoperative analgesia. Our primary analysis was noninferiority of serratus anterior blocks compared with paravertebral blocks. RESULTS: Baseline characteristics were comparable among the three groups. Two hours after surgery, the mean difference in visual analogue pain scores between the serratus anterior and paravertebral blocks was 0.0 (96.8% CI -0.4 to 0.3) cm at rest, -0.2 (-0.8 to 0.4) cm during coughing and -0.1(-0.5 to 0.3) for Prince-Henry pain scores. After 24 h, the mean difference was 0.0 (-0.7 to 0.8) cm at rest, 0.1 (-0.8 to 0.9) cm during coughing and 0.1(-0.4 to 0.6) for Prince-Henry pain scores. All differences were significantly noninferior. Time to onset of pain after surgery was 19 ±â€Š5 (SD) hours with serratus anterior blocks, 16 ±â€Š5 h with paravertebral blocks and 12 ±â€Š5 h with general anaesthesia. Anaesthesia with either block was associated with significantly less intra-operative propofol and sufentanil, reduced postoperative rescue analgesia (tramadol) and less postoperative nausea and vomiting compared with general anaesthesia alone. Patients with serratus anterior block had a significantly lower incidence of intra-operative hypotension and requirement for intra-operative vasopressor (3.4%), compared with general anaesthesia alone. Serratus anterior block took less time to perform than paravertebral block (5.1 ±â€Š1.1 min versus 10.1 ±â€Š2.9 min). CONCLUSION: Serratus anterior plane blocks, which are easier and quicker than paravertebral blocks, provide comparable analgesia in patients having video-assisted thoracic surgery. CLINICAL TRIAL NUMBER AND REGISTRY URL: ChiCTR1800017671; http://www.chictr.org.cn/hvshowproject.aspx?id=13510.

3.
J Biol Chem ; 289(7): 4262-72, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24375408

RESUMEN

Transient Receptor Potential mucolipin (TRPML) channels are implicated in endolysosomal trafficking, lysosomal Ca(2+) and Fe(2+) release, lysosomal biogenesis, and autophagy. Mutations in human TRPML1 cause the lysosome storage disease, mucolipidosis type IV (MLIV). Unlike vertebrates, which express three TRPML genes, TRPML1-3, the Drosophila genome encodes a single trpml gene. Although the trpml-deficient flies exhibit cellular defects similar to those in mammalian TRPML1 mutants, the biophysical properties of Drosophila TRPML channel remained uncharacterized. Here, we show that transgenic expression of human TRPML1 in the neurons of Drosophila trpml mutants partially suppressed the pupal lethality phenotype. When expressed in HEK293 cells, Drosophila TRPML was localized in both endolysosomes and plasma membrane and was activated by phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2) applied to the cytoplasmic side in whole lysosomes and inside-out patches excised from plasma membrane. The PI(3,5)P2-evoked currents were blocked by phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), but not other phosphoinositides. Using TRPML A487P, which mimics the varitint-waddler (Va) mutant of mouse TRPML3 with constitutive whole-cell currents, we show that TRPML is biphasically regulated by extracytosolic pH, with an optimal pH about 0.6 pH unit higher than that of human TRPML1. In addition to monovalent cations, TRPML exhibits high permeability to Ca(2+), Mn(2+), and Fe(2+), but not Fe(3+). The TRPML currents were inhibited by trivalent cations Fe(3+), La(3+), and Gd(3+). These features resemble more closely to mammalian TRPML1 than TRPML2 and TRPML3, but with some obvious differences. Together, our data support the use of Drosophila for assessing functional significance of TRPML1 in cell physiology.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de Drosophila/metabolismo , Endosomas/metabolismo , Lisosomas/metabolismo , Metales/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Cationes/metabolismo , Membrana Celular/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Endosomas/genética , Células HEK293 , Humanos , Transporte Iónico/fisiología , Lisosomas/genética , Mutación Missense , Fosfatos de Fosfatidilinositol/genética , Canales de Potencial de Receptor Transitorio/genética
4.
Pflugers Arch ; 466(7): 1301-16, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24121765

RESUMEN

In the central nervous system, canonical transient receptor potential (TRPC) channels have been implicated in mediating neuronal excitation induced by stimulating metabotropic receptors, including group 1 metabotropic glutamate receptors (mGluRs). Lateral septal (LS) neurons express high levels of TRPC4 and group I mGluRs. However, to what extent native TRPC4-containing channels (TRPC4-cc) are activated as well as the impact of different levels of TRPC4-cc activation on neuronal excitability remain elusive. Here, we report that stimulating LS neurons with group I mGluR agonist, (S)-3,5-DHPG, causes either an immediate increase in firing rate or an initial burst followed by a pause of firing, which can be correlated with below-threshold-depolarization (BTD) or above-threshold-plateau-depolarization (ATPD), respectively, in whole-cell recordings. The early phase of BTD and the entire ATPD are completely absent in neurons from TRPC4−/− mice. Moreover, in the same LS neurons, BTD can be converted to ATPD at more depolarized potentials or with a brief current injection, suggesting that BTD and ATPD may represent partial and full activations of TRPC4-cc, respectively. We show that coincident mGluR stimulation and depolarization is required to evoke strong TRPC4-cc current, and Na+ and Ca2+ influx, together with dynamic changes of intracellular Ca(2+), are essential for ATPD induction. Our results suggest that TRPC4-cc integrates metabotropic receptor stimulation with intracellular Ca(2+) signals to generate two interconvertible depolarization responses to affect excitability of LS neurons in distinct fashions.


Asunto(s)
Potenciales de Acción , Neuronas/metabolismo , Núcleos Septales/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Agonistas de Aminoácidos Excitadores/farmacología , Glicina/análogos & derivados , Glicina/farmacología , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/fisiología , Receptor del Glutamato Metabotropico 5/metabolismo , Resorcinoles/farmacología , Núcleos Septales/citología , Sodio/metabolismo , Canales Catiónicos TRPC/genética
5.
Pflugers Arch ; 466(7): 1273-87, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24092046

RESUMEN

Transient receptor potential A1 (TRPA1) is implicated in somatosensory processing and pathological pain sensation. Although not strictly voltage-gated, ionic currents of TRPA1 typically rectify outwardly, indicating channel activation at depolarized membrane potentials. However, some reports also showed TRPA1 inactivation at high positive potentials, implicating voltage-dependent inactivation. Here we report a conserved leucine residue, L906, in the putative pore helix, which strongly impacts the voltage dependency of TRPA1. Mutation of the leucine to cysteine (L906C) converted the channel from outward to inward rectification independent of divalent cations and irrespective to stimulation by allyl isothiocyanate. The mutant, but not the wild-type channel, displayed exclusively voltage-dependent inactivation at positive potentials. The L906C mutation also exhibited reduced sensitivity to inhibition by TRPA1 blockers, HC030031 and ruthenium red. Further mutagenesis of the leucine to all natural amino acids individually revealed that most substitutions at L906 (15/19) resulted in inward rectification, with exceptions of three amino acids that dramatically reduced channel activity and one, methionine, which mimicked the wild-type channel. Our data are plausibly explained by a bimodal gating model involving both voltage-dependent activation and inactivation of TRPA1. We propose that the key pore helix residue, L906, plays an essential role in responding to the voltage-dependent gating.


Asunto(s)
Activación del Canal Iónico , Mutación Missense , Canales de Potencial de Receptor Transitorio/metabolismo , Secuencia de Aminoácidos , Animales , Células HEK293 , Humanos , Leucina/genética , Potenciales de la Membrana , Ratones , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/química , Canales de Potencial de Receptor Transitorio/genética
6.
bioRxiv ; 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38585789

RESUMEN

The transcription repressor REST in the dorsal root ganglion (DRG) is upregulated by peripheral nerve injury and promotes the development of chronic pain. However, the genes targeted by REST in neuropathic pain development remain unclear. The expression levels of 4 opioid receptor (Oprm1, Oprd1, Oprl1, Oprk1) and the cannabinoid CB1 receptor (Cnr1) genes in the DRG regulate nociception. In this study, we determined the role of REST in the control of their expression in the DRG induced by spared nerve injury (SNI) in both male and female mice. Transcriptomic analyses of male mouse DRGs followed by quantitative reverse transcription polymerase chain reaction analyses of both male and female mouse DRGs showed that SNI upregulated expression of Rest and downregulated mRNA levels of all 4 opioid receptor and Cnr1 genes, but Oprm1 was upregulated in female mice. Analysis of publicly available bioinformatic data suggested that REST binds to the promoter regions of Oprm1 and Cnr1. Chromatin immunoprecipitation analyses indicated differing levels of REST at these promoters in male and female mice. Full-length Rest conditional knockout in primary sensory neurons reduced SNI-induced pain hypersensitivity and rescued the SNI-induced reduction in the expression of Oprd1 and Cnr1 in the DRG in both male and female mice. Our results suggest that nerve injury represses the transcription of Oprd1 and Cnr1 via REST in primary sensory neurons and that REST is a potential therapeutic target for neuropathic pain.

7.
Mol Pharmacol ; 84(4): 511-20, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23847084

RESUMEN

Chronic pain is thought to be partly caused by a loss of GABAergic inhibition and resultant neuronal hyperactivation in the central pain-modulating system, but the underlying mechanisms for pain-modulating neurons in the brain are unclear. In this study, we investigated the cellular mechanisms for activation of brainstem descending pain facilitation in rats under persistent pain conditions. In the nucleus raphe magnus (NRM), a critical relay in the brain's descending pain-modulating system, persistent inflammatory pain induced by complete Freund's adjuvant decreased the protein level of K(+)-Cl(-) cotransporter (KCC2) in both total and synaptosomal preparations. Persistent pain also shifted the equilibrium potential of GABAergic inhibitory postsynaptic current (EIPSC) to a more positive level and increased the firing of evoked action potentials selectively in µ-opioid receptor (MOR)-expressing NRM neurons, but not in MOR-lacking NRM neurons. Microinjection of brain-derived neurotrophic factor (BDNF) into the NRM inhibited the KCC2 protein level in the NRM, and both BDNF administration and KCC2 inhibition by furosemide mimicked the pain-induced effects on EIPSC and excitability in MOR-expressing neurons. Furthermore, inhibiting BDNF signaling by NRM infusion of tyrosine receptor kinase B-IgG or blocking KCC2 with furosemide prevented these pain effects in MOR-expressing neurons. These findings demonstrate a cellular mechanism by which the hyperactivity of NRM MOR-expressing neurons, presumably responsible for descending pain facilitation, contributes to pain sensitization through the signaling cascade of BDNF-KCC2-GABA impairment in the development of chronic pain.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Regulación hacia Abajo/fisiología , Dolor/metabolismo , Núcleos del Rafe/metabolismo , Simportadores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Animales Recién Nacidos , Tronco Encefálico/citología , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/metabolismo , Factor Neurotrófico Derivado del Encéfalo/fisiología , Regulación hacia Abajo/efectos de los fármacos , Masculino , Microinyecciones , Técnicas de Cultivo de Órganos , Núcleos del Rafe/citología , Núcleos del Rafe/efectos de los fármacos , Ratas , Ratas Wistar , Simportadores/antagonistas & inhibidores , Cotransportadores de K Cl
8.
Neuron ; 111(13): 2038-2050.e6, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37146610

RESUMEN

Neuropathic pain is a common, debilitating chronic pain condition caused by damage or a disease affecting the somatosensory nervous system. Understanding the pathophysiological mechanisms underlying neuropathic pain is critical for developing new therapeutic strategies to treat chronic pain effectively. Tiam1 is a Rac1 guanine nucleotide exchange factor (GEF) that promotes dendritic and synaptic growth during hippocampal development by inducing actin cytoskeletal remodeling. Here, using multiple neuropathic pain animal models, we show that Tiam1 coordinates synaptic structural and functional plasticity in the spinal dorsal horn via actin cytoskeleton reorganization and synaptic NMDAR stabilization and that these actions are essential for the initiation, transition, and maintenance of neuropathic pain. Furthermore, an antisense oligonucleotides (ASO) targeting spinal Tiam1 persistently alleviate neuropathic pain sensitivity. Our findings suggest that Tiam1-coordinated synaptic functional and structural plasticity underlies the pathophysiology of neuropathic pain and that intervention of Tiam1-mediated maladaptive synaptic plasticity has long-lasting consequences in neuropathic pain management.


Asunto(s)
Dolor Crónico , Neuralgia , Animales , Factores de Intercambio de Guanina Nucleótido/genética , Plasticidad Neuronal/fisiología , Actinas , Neuralgia/terapia
9.
J Clin Invest ; 132(24)2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36519542

RESUMEN

Chronic pain often leads to depression, increasing patient suffering and worsening prognosis. While hyperactivity of the anterior cingulate cortex (ACC) appears to be critically involved, the molecular mechanisms underlying comorbid depressive symptoms in chronic pain remain elusive. T cell lymphoma invasion and metastasis 1 (Tiam1) is a Rac1 guanine nucleotide exchange factor (GEF) that promotes dendrite, spine, and synapse development during brain development. Here, we show that Tiam1 orchestrates synaptic structural and functional plasticity in ACC neurons via actin cytoskeleton reorganization and synaptic N-methyl-d-aspartate receptor (NMDAR) stabilization. This Tiam1-coordinated synaptic plasticity underpins ACC hyperactivity and drives chronic pain-induced depressive-like behaviors. Notably, administration of low-dose ketamine, an NMDAR antagonist emerging as a promising treatment for chronic pain and depression, induces sustained antidepressant-like effects in mouse models of chronic pain by blocking Tiam1-mediated maladaptive synaptic plasticity in ACC neurons. Our results reveal Tiam1 as a critical factor in the pathophysiology of chronic pain-induced depressive-like behaviors and the sustained antidepressant-like effects of ketamine.


Asunto(s)
Dolor Crónico , Ketamina , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Animales , Ratones , Antidepresivos/farmacología , Dolor Crónico/tratamiento farmacológico , Depresión/tratamiento farmacológico , Depresión/genética , Ketamina/farmacología , Plasticidad Neuronal , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/genética , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/metabolismo
10.
J Gen Physiol ; 153(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33320167

RESUMEN

TRPV3 is a temperature-sensitive, nonselective cation channel expressed prominently in skin keratinocytes. TRPV3 plays important roles in hair morphogenesis and maintenance of epidermal barrier function. Gain-of-function mutations of TRPV3 have been found in both humans and rodents and are associated with hair loss, pruritus, and dermatitis. Here, we study the mechanisms of acid regulation of TRPV3 by using site-directed mutagenesis, fluorescent intracellular calcium measurement, and whole-cell patch-clamp recording techniques. We show that, whereas extracellular acid inhibits agonist-induced TRPV3 activation through an aspartate residue (D641) in the selectivity filter, intracellular protons sensitize the channel through cytoplasmic C-terminal glutamate and aspartate residues (E682, E689, and D727). Neutralization of the three C-terminal residues presensitizes the channel to agonist stimulation. Molecular dynamic simulations revealed that charge neutralization of the three C-terminal residues stabilized the sensitized channel conformation and enhanced the probability of α-helix formation in the linker between the S6 transmembrane segment and TRP domain. We conclude that acid inhibits TRPV3 function from the extracellular side but facilitates it from the intracellular side. These novel mechanisms of TRPV3 proton sensing can offer new insights into the role of TRPV3 in the regulation of epidermal barrier permeability and skin disorders under conditions of tissue acidosis.


Asunto(s)
Protones , Canales Catiónicos TRPV , Queratinocitos , Técnicas de Placa-Clamp , Piel , Canales Catiónicos TRPV/genética
11.
Brain Res Bull ; 169: 18-24, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33400956

RESUMEN

Resveratrol, a naturally occurring stilbene found in red wine, is known to modulate the activity of several types of ion channels and membrane receptors, including Ca2+, K+, and Na+ ion channels. However, little is known about the effects of resveratrol on some important receptors, such as glycine receptors and GABAA receptors, in the central nervous system (CNS). In the present study, the effects of resveratrol on glycine receptor or GABAA receptor-mediated currents in cultured rat inferior colliculus (IC) and auditory cortex (AC) neurons were studied using whole-cell voltage-clamp recordings. Resveratrol itself did not evoke any currents in IC neurons but it reversibly decreased the amplitude of glycine-induced current (IGly) in a concentration-dependent manner. Resveratrol did not change the reversal potential of IGly but it shifted the concentration-response relationship to the right without changing the Hill coefficient and with decreasing the maximum response of IGly. Interestingly, resveratrol inhibited the amplitude of IGly but not that of GABA-induced current (IGABA) in AC neurons. More importantly, resveratrol inhibited GlyR-mediated but not GABAAR-mediated inhibitory postsynaptic currents in IC neurons using brain slice recordings. Together, these results demonstrate that resveratrol noncompetitively inhibits IGly in auditory neurons by decreasing the affinity of glycine to its receptor. These findings suggest that the native glycine receptors but not GABAA receptors in central neurons are targets of resveratrol during clinical administrations.


Asunto(s)
Colículos Inferiores/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores de Glicina/metabolismo , Resveratrol/farmacología , Transmisión Sináptica/efectos de los fármacos , Animales , Colículos Inferiores/metabolismo , Neuronas/metabolismo , Técnicas de Placa-Clamp , Ratas
12.
Nat Commun ; 12(1): 2662, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976218

RESUMEN

Central leptin action rescues type 1 diabetic (T1D) hyperglycemia; however, the underlying mechanism and the identity of mediating neurons remain elusive. Here, we show that leptin receptor (LepR)-expressing neurons in arcuate (LepRArc) are selectively activated in T1D. Activation of LepRArc neurons, Arc GABAergic (GABAArc) neurons, or arcuate AgRP neurons, is able to reverse the leptin's rescuing effect. Conversely, inhibition of GABAArc neurons, but not AgRP neurons, produces leptin-mimicking rescuing effects. Further, AgRP neuron function is not required for T1D hyperglycemia or leptin's rescuing effects. Finally, T1D LepRArc neurons show defective nutrient sensing and signs of cellular energy deprivation, which are both restored by leptin, whereas nutrient deprivation reverses the leptin action. Our results identify aberrant activation of LepRArc neurons owing to energy deprivation as the neural basis for T1D hyperglycemia and that leptin action is mediated by inhibiting LepRArc neurons through reversing energy deprivation.


Asunto(s)
Encéfalo/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Hiperglucemia/metabolismo , Leptina/metabolismo , Neuronas/metabolismo , Receptores de Leptina/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Glucemia/metabolismo , Encéfalo/citología , Encéfalo/efectos de los fármacos , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/sangre , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Infusiones Intraventriculares , Leptina/administración & dosificación , Masculino , Ratones Transgénicos , Neuronas/efectos de los fármacos , Receptores de Leptina/genética , Transducción de Señal/efectos de los fármacos
13.
Mol Cancer Ther ; 20(9): 1680-1691, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34158347

RESUMEN

We analyzed the efficacy and mechanistic interactions of PARP inhibition (PARPi; olaparib) and CDK4/6 inhibition (CDK4/6i; palbociclib or abemaciclib) combination therapy in castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC) models. We demonstrated that combined olaparib and palbociblib or abemaciclib treatment resulted in synergistic suppression of the p-Rb1-E2F1 signaling axis at the transcriptional and posttranslational levels, leading to disruption of cell-cycle progression and inhibition of E2F1 gene targets, including genes involved in DDR signaling/damage repair, antiapoptotic BCL-2 family members (BCL-2 and MCL-1), CDK1, and neuroendocrine differentiation (NED) markers in vitro and in vivo In addition, olaparib + palbociclib or olaparib + abemaciclib combination treatment resulted in significantly greater growth inhibition and apoptosis than either single agent alone. We further showed that PARPi and CDK4/6i combination treatment-induced CDK1 inhibition suppressed p-S70-BCL-2 and increased caspase cleavage, while CDK1 overexpression effectively prevented the downregulation of p-S70-BCL-2 and largely rescued the combination treatment-induced cytotoxicity. Our study defines a novel combination treatment strategy for CRPC and NEPC and demonstrates that combination PARPi and CDK4/6i synergistically promotes suppression of the p-Rb1-E2F1 axis and E2F1 target genes, including CDK1 and NED proteins, leading to growth inhibition and increased apoptosis in vitro and in vivo Taken together, our results provide a molecular rationale for PARPi and CDK4/6i combination therapy and reveal mechanism-based clinical trial opportunities for men with NEPC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Diferenciación Celular , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Tumores Neuroectodérmicos/tratamiento farmacológico , Poli(ADP-Ribosa) Polimerasas/química , Neoplasias de la Próstata/tratamiento farmacológico , Aminopiridinas/administración & dosificación , Animales , Apoptosis , Bencimidazoles/administración & dosificación , Ciclo Celular , Proliferación Celular , Humanos , Masculino , Ratones , Ratones Desnudos , Tumores Neuroectodérmicos/metabolismo , Tumores Neuroectodérmicos/patología , Ftalazinas/administración & dosificación , Piperazinas/administración & dosificación , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Piridinas/administración & dosificación , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Clin Exp Pharmacol Physiol ; 37(4): 435-40, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19878215

RESUMEN

1. The diuretic amiloride is known to modulate the activity of several types of ion channels and membrane receptors in addition to its inhibitory effects on many ion transport systems. However, the effects of amiloride on some important ion channels and receptors, such as GABA(A) receptors, in the central nervous system have not been characterized. 2. In the present study, we investigated the functional action of amiloride on native GABA(A) receptors in cultured neurons of rat inferior colliculus using whole-cell patch-clamp recordings. 3. Amiloride reversibly inhibited the amplitude of the GABA-induced current (I(GABA)) in a concentration-dependent manner (IC(50) 454 +/- 24 micromol/L) under conditions of voltage-clamp with a holding potential at -60 mV. The inhibition depended on drug application mode and was independent of membrane potential. Amiloride did not change the reversal potential of I(GABA). Moreover, amiloride induced a parallel right-ward shift in the concentration-response curve for I(GABA) without altering the maximal value and Hill coefficient. 4. The present study shows that amiloride competitively inhibits the current mediated by native GABA(A) receptors in the brain region, probably via a direct action on GABA-binding sites on the receptor. The findings suggest that the functional actions of amiloride on GABA(A) receptors may result in possible side-effects on the central nervous system in the case of direct application of this drug into the cerebrospinal fluid for treatment of diseases such as brain tumours.


Asunto(s)
Amilorida/farmacología , Diuréticos/farmacología , Antagonistas del GABA/farmacología , Antagonistas de Receptores de GABA-A , Colículos Inferiores/efectos de los fármacos , Neuronas/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos , Amilorida/efectos adversos , Animales , Animales Recién Nacidos , Células Cultivadas , Diuréticos/efectos adversos , Antagonistas del GABA/efectos adversos , Colículos Inferiores/citología , Cinética , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Bloqueadores de los Canales de Sodio/efectos adversos , Bloqueadores de los Canales de Sodio/farmacología
15.
Brain Res ; 1748: 147071, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32827550

RESUMEN

GABAA receptors (GABAARs) and glycine receptors (GlyRs) are two principal inhibitory chloride ion channels in the central nervous system. The two receptors do not function independently but cross-talk to each other, i.e., the activation of one receptor would inhibit the other. This cross-talk is present in different patterns across various regions in the central nervous system; however, the factor that determines these patterns is not understood. Here, we show that the pattern of cross-talk between the two receptors is shaped by their relative expression level in a neuron: a higher expression level correlates with louder talk. In line with a tendency of decrease in expression level of GlyRs and increase in expression level of GABAARs from the spinal cord, the brainstem to the neocortex, GlyRs talked much louder (i.e. produced greater inhibition) than GABAARs (one-way pattern) in spinal cord neurons, about equally loud as GABAARs (symmetric pattern) in inferior colliculus neurons and less loud (i.e. less inhibition) than GABAARs (asymmetric pattern) in auditory cortex neurons. Overexpression of GlyRs in inferior colliculus neurons produced an asymmetric pattern that should otherwise have been observed in spinal cord neurons. These expression level-dependent patterns of cross-talk between the two receptors may suggest how the central nervous system uses an alternative mechanism to maintain a delicate level of inhibition through adjusting the proportion of the two receptors in a neuron along its pathway.


Asunto(s)
Neuronas/metabolismo , Receptores de GABA-A/metabolismo , Receptores de Glicina/metabolismo , Médula Espinal/metabolismo , Animales , Corteza Auditiva/metabolismo , Células Cultivadas , Colículos Inferiores/metabolismo , Técnicas de Placa-Clamp , Ratas
16.
Cells ; 9(3)2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-32110987

RESUMEN

Phospholipid scramblase 1 (PLSCR1), a lipid-binding and Ca2+-sensitive protein located on plasma membranes, is critically involved in phosphatidylserine (PS) externalization, an important process in cell apoptosis. Transient receptor potential canonical 5 (TRPC5), is a nonselective Ca2+ channel in neurons that interacts with many downstream molecules, participating in diverse physiological functions including temperature or mechanical sensation. The interaction between TRPC5 and PLSCR1 has never been reported. Here, we showed that PLSCR1 interacts with TRPC5 through their C-termini in HEK293 cells and mouse cortical neurons. Formation of TRPC5-PLSCR1 complex stimulates PS externalization and promotes cell apoptosis in HEK293 cells and mouse cerebral neurons. Furthermore, in vivo studies showed that PS externalization in cortical neurons induced by artificial cerebral ischemia-reperfusion was reduced in TRPC5 knockout mice compared to wild-type mice, and that the percentage of apoptotic neurons was also lower in TRPC5 knockout mice than in wild-type mice. Collectively, the present study suggested that TRPC5-PLSCR1 is a signaling complex mediating PS externalization and apoptosis in neurons and that TRPC5 plays a pathological role in cerebral-ischemia reperfusion injury.


Asunto(s)
Apoptosis , Exocitosis , Neuronas/metabolismo , Fosfatidilserinas/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Membrana Celular/metabolismo , Células HEK293 , Humanos , Activación del Canal Iónico , Ratones Noqueados , Unión Proteica , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Transducción de Señal
17.
Nat Commun ; 11(1): 3794, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32732906

RESUMEN

Defective rhythmic metabolism is associated with high-fat high-caloric diet (HFD) feeding, ageing and obesity; however, the neural basis underlying HFD effects on diurnal metabolism remains elusive. Here we show that deletion of BMAL1, a core clock gene, in paraventricular hypothalamic (PVH) neurons reduces diurnal rhythmicity in metabolism, causes obesity and diminishes PVH neuron activation in response to fast-refeeding. Animal models mimicking deficiency in PVH neuron responsiveness, achieved through clamping PVH neuron activity at high or low levels, both show obesity and reduced diurnal rhythmicity in metabolism. Interestingly, the PVH exhibits BMAL1-controlled rhythmic expression of GABA-A receptor γ2 subunit, and dampening rhythmicity of GABAergic input to the PVH reduces diurnal rhythmicity in metabolism and causes obesity. Finally, BMAL1 deletion blunts PVH neuron responses to external stressors, an effect mimicked by HFD feeding. Thus, BMAL1-driven PVH neuron responsiveness in dynamic activity changes involving rhythmic GABAergic neurotransmission mediates diurnal rhythmicity in metabolism and is implicated in diet-induced obesity.


Asunto(s)
Factores de Transcripción ARNTL/genética , Ritmo Circadiano/fisiología , Obesidad/patología , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de GABA-A/metabolismo , Animales , Ritmo Circadiano/genética , Dieta Alta en Grasa , Metabolismo Energético/fisiología , Conducta Alimentaria/fisiología , Ratones , Ratones Noqueados , Neuronas/fisiología , Obesidad/genética , Núcleo Hipotalámico Paraventricular/citología
18.
eNeuro ; 6(4)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31331938

RESUMEN

The paraventricular hypothalamus (PVH) regulates stress, feeding behaviors and other homeostatic processes, but whether PVH also drives defensive states remains unknown. Here we showed that photostimulation of PVH neurons in mice elicited escape jumping, a typical defensive behavior. We mapped PVH outputs that densely terminate in the ventral midbrain (vMB) area, and found that activation of the PVH→vMB circuit produced profound defensive behavioral changes, including escape jumping, hiding, hyperlocomotion, and learned aversion. Electrophysiological recordings showed excitatory postsynaptic input onto vMB neurons via PVH fiber activation, and in vivo studies demonstrated that glutamate transmission from PVH→vMB was required for the evoked behavioral responses. Photostimulation of PVH→vMB fibers induced cFos expression mainly in non-dopaminergic neurons. Using a dual optogenetic-chemogenetic strategy, we further revealed that escape jumping and hiding were partially contributed by the activation of midbrain glutamatergic neurons. Taken together, our work unveils a hypothalamic-vMB circuit that encodes defensive properties, which may be implicated in stress-induced defensive responses.


Asunto(s)
Reacción de Fuga/fisiología , Mesencéfalo/fisiología , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Animales , Reacción de Prevención/fisiología , Conducta Animal , Ingestión de Alimentos/fisiología , Ácido Glutámico/fisiología , Masculino , Mesencéfalo/citología , Ratones Transgénicos , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Optogenética , Núcleo Hipotalámico Paraventricular/citología
19.
Nat Commun ; 10(1): 3446, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31371721

RESUMEN

Feeding is known to be profoundly affected by stress-related emotional states and eating disorders are comorbid with psychiatric symptoms and altered emotional responses. The neural basis underlying feeding regulation by stress-related emotional changes is poorly understood. Here, we identify a novel projection from the paraventricular hypothalamus (PVH) to the ventral lateral septum (LSv) that shows a scalable regulation on feeding and behavioral changes related to emotion. Weak photostimulation of glutamatergic PVH→LSv terminals elicits stress-related self-grooming and strong photostimulation causes fear-related escape jumping associated with respective weak and strong inhibition on feeding. In contrast, inhibition of glutamatergic inputs to LSv increases feeding with signs of reduced anxiety. LSv-projecting neurons are concentrated in rostral PVH. LSv and LSv-projecting PVH neurons are activated by stressors in vivo, whereas feeding bouts were associated with reduced activity of these neurons. Thus, PVH→LSv neurotransmission underlies dynamic feeding by orchestrating emotional states, providing a novel neural circuit substrate underlying comorbidity between eating abnormalities and psychiatric disorders.


Asunto(s)
Conducta Alimentaria/fisiología , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Distrés Psicológico , Animales , Conducta Animal , Fármacos actuantes sobre Aminoácidos Excitadores , Trastornos de Alimentación y de la Ingestión de Alimentos , Aseo Animal/fisiología , Masculino , Ratones , Modelos Animales , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
20.
Sci Adv ; 5(3): eaav1640, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30854429

RESUMEN

Animals must consider competing information before deciding to eat: internal signals indicating the desirability of food and external signals indicating the risk involved in eating within a particular environment. The behaviors driven by the former are manifestations of hunger, and the latter, anxiety. The connection between pathologic anxiety and reduced eating in conditions like typical depression and anorexia is well known. Conversely, anti-anxiety drugs such as benzodiazepines increase appetite. Here, we show that GABAergic neurons in the diagonal band of Broca (DBBGABA) are responsive to indications of risk and receive monosynaptic inhibitory input from lateral hypothalamus GABAergic neurons (LHGABA). Activation of this circuit reduces anxiety and causes indiscriminate feeding. We also found that diazepam rapidly reduces DBBGABA activity while inducing indiscriminate feeding. Our study reveals that the LHGABA→DBBGABA neurocircuit overrides anxiogenic environmental cues to allow feeding and that this pathway may underlie the link between eating and anxiety-related disorders.


Asunto(s)
Prosencéfalo Basal/fisiología , Señales (Psicología) , Ambiente , Conducta Alimentaria , Área Hipotalámica Lateral/fisiología , Red Nerviosa , Animales , Ansiedad , Neuronas GABAérgicas/fisiología , Ratones , Transmisión Sináptica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA