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










Base de datos
Intervalo de año de publicación
1.
Neuropharmacology ; 238: 109654, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37437688

RESUMEN

The N-methyl-d-aspartate receptor (NMDAR) is an enigmatic macromolecule that has garnered a good deal of attention on account of its involvement in the cellular processes that underlie learning and memory, following its discovery in the mid twentieth century (Baudry and Davis, 1991). Yet, despite advances in knowledge about its function, there remains much more to be uncovered regarding the receptor's biophysical properties, subunit composition, and role in CNS physiology and pathophysiology. The motivation for this review stems from the need for synthesizing new information gathered about these receptors that sheds light on their role in synaptic plasticity and their dichotomous relationship with the amino acid d-serine through which they influence the pathogenesis of neurodegenerative diseases like temporal lobe epilepsy (TLE), the most common type of adult epilepsies (Beesley et al., 2020a). This review will outline pertinent ideas relating structure and function of t-NMDARs (GluN3 subunit-containing triheteromeric NMDARs) for which d-serine might serve as an inverse co-agonist. We will explore how tracing d-serine's origins blends glutamate-receptor biology with glial biology to help provide fresh perspectives on how neurodegeneration might interlink with neuroinflammation to initiate and perpetuate the disease state. Taken together, we envisage the review to deepen our understanding of endogenous d-serine's new role in the brain while also recognizing its therapeutic potential in the treatment of TLE that is oftentimes refractory to medications.


Asunto(s)
Epilepsia del Lóbulo Temporal , Receptores de N-Metil-D-Aspartato , Humanos , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Encéfalo/metabolismo
2.
Front Synaptic Neurosci ; 15: 1156777, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37292368

RESUMEN

N-methyl-D-aspartate receptors (NMDARs) are one of three ligand-gated ionotropic channels that transduce the effects of neurotransmitter glutamate at excitatory synapses within the central nervous system. Their ability to influx Ca2+ into cells, unlike mature AMPA or kainate receptors, implicates them in a variety of processes ranging from synaptic plasticity to cell death. Many of the receptor's capabilities, including binding glutamate and regulating Ca2+ influx, have been attributed to their subunit composition, determined putatively using cell biology, electrophysiology and/or pharmacology. Here, we show that subunit composition of synaptic NMDARs can also be readily visualized in acute brain slices (rat) using highly specific antibodies directed against extracellular epitopes of the subunit proteins and high-resolution confocal microscopy. This has helped confirm the expression of triheteromeric t-NMDARs (containing GluN1, GluN2, and GluN3 subunits) at synapses for the first time and reconcile functional differences with diheteromeric d-NMDARs (containing GluN1 and GluN2 subunits) described previously. Even though structural information about individual receptors is still diffraction limited, fluorescently tagged receptor subunit puncta coalesce with precision at various magnifications and/or with the postsynaptic density (PSD-95) but not the presynaptic active zone marker Bassoon. These data are particularly relevant for identifying GluN3A-containing t-NMDARs that are highly Ca2+ permeable and whose expression at excitatory synapses renders neurons vulnerable to excitotoxicity and cell death. Imaging NMDAR subunit proteins at synapses not only offers firsthand insights into subunit composition to correlate function but may also help identify zones of vulnerability within brain structures underlying neurodegenerative diseases like Temporal Lobe Epilepsy.

3.
J Neurophysiol ; 127(6): 1496-1510, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35475675

RESUMEN

Temporal lobe epilepsy (TLE) is the most common type of epilepsy in adults that is often refractory to antiepileptic medication therapy. Neither the pathology nor the etiology of TLE is fully characterized, although recent studies have established that the two are causally related. TLE pathology entails a stereotypic pattern of neuron loss in hippocampal and parahippocampal regions, predominantly in CA1 subfield of the hippocampus and layer 3 of the medial entorhinal area (MEA), deemed hallmark pathological features of the disease. Through this work, we address the contribution of glutamatergic N-methyl-d-aspartate receptors (NMDARs) to the pathology (vulnerability and pattern of neuronal loss), and by extension to the pathophysiology (Ca2+-induced excitotoxicity), by assaying the spatial expression of their subunit proteins (GluN1, GluN2A, GluN2B, and GluN3A) in these regions using area-specific tissue analysis (ASTA), a novel methodology for harvesting brain chads from hard-to-reach regions within brain slices for Western blotting. Our data suggest gradient expression of the GluN3A subunit along the mid-lateral extent of layer 3 MEA and along the CA1-subicular axis in the hippocampus, unlike GluN1 or GluN2 subunits that are uniformly distributed. Incorporation of GluN3A in the subunit composition of conventional diheteromeric (d-) NMDARs yield triheteromeric (t-) NMDARs which by virtue of their increased selectivity for Ca2+ render neurons vulnerable to excitotoxic damage. Thus, the expression profile of this subunit sheds light on the spatial extent of the pathology observed in these regions and implicates the GluN3 subunit of NMDARs in hippocampal and entorhinal cortical pathology underlying TLE.NEW & NOTEWORTHY The role of the GluN3 subunit in NMDAR-mediated pathophysiology underlying TLE is not known. Here, we demonstrate using ASTA (area-specific tissue analysis) that its expression in specific regions of the entorhinal cortex and the hippocampus is correlated with significant cell loss and neurodegeneration, hallmark features of the disease.


Asunto(s)
Corteza Entorrinal , Epilepsia del Lóbulo Temporal , Epilepsia , Glicoproteínas de Membrana , Receptores de N-Metil-D-Aspartato , Animales , Modelos Animales de Enfermedad , Corteza Entorrinal/metabolismo , Corteza Entorrinal/patología , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Glicoproteínas de Membrana/metabolismo , Neuronas/metabolismo , Ratas , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
4.
Neuroscience ; 453: 168-186, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33197499

RESUMEN

Entrainment of the hippocampus by the medial entorhinal area (MEA) in Temporal Lobe Epilepsy (TLE), the most common type of drug-resistant epilepsy in adults, is believed to be mediated primarily through the perforant pathway (PP), which connects stellate cells in layer (L) II of the MEA with granule cells of the dentate gyrus (DG) to drive the hippocampal tri-synaptic circuit. Using immunohistochemistry, high-resolution confocal microscopy and the rat pilocarpine model of TLE, we show here that the lesser known temporoammonic pathway (TAP) plays a significant role in transferring MEA pathology to the CA1 region of the hippocampus independently of the PP. The pathology observed was region-specific and restricted primarily to the CA1c subfield of the hippocampus. As shown previously, daily intracranial infusion of d-serine (100 µm), an antagonist of GluN3-containing triheteromeric N-Methyl d-aspartate receptors (t-NMDARs), into the MEA prevented loss of LIII neurons and epileptogenesis. This intervention in the MEA led to the rescue of hippocampal CA1 neurons that would have otherwise perished in the epileptic animals, and down regulation of the expression of astrocytes and microglia thereby mitigating the effects of neuroinflammation. Interestingly, these changes were not observed to a similar extent in other regions of vulnerability like the hilus, DG or CA3, suggesting that the pathology manifest in CA1 is driven predominantly through the TAP. This work highlights TAP's role in the entrainment of the hippocampus and identifies specific areas for therapeutic intervention in dealing with TLE.


Asunto(s)
Epilepsia del Lóbulo Temporal , Animales , Corteza Entorrinal , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Hipocampo , Pilocarpina , Ratas , Serina
5.
Nat Commun ; 11(1): 4966, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33009404

RESUMEN

Temporal lobe epilepsy (TLE) is the most common type of drug-resistant epilepsy in adults, with an unknown etiology. A hallmark of TLE is the characteristic loss of layer 3 neurons in the medial entorhinal area (MEA) that underlies seizure development. One approach to intervention is preventing loss of these neurons through better understanding of underlying pathophysiological mechanisms. Here, we show that both neurons and glia together give rise to the pathology that is mitigated by the amino acid D-serine whose levels are potentially diminished under epileptic conditions. Focal administration of D-serine to the MEA attenuates neuronal loss in this region thereby preventing epileptogenesis in an animal model of TLE. Additionally, treatment with D-serine reduces astrocyte counts in the MEA, alters their reactive status, and attenuates proliferation and/or infiltration of microglia to the region thereby curtailing the deleterious consequences of neuroinflammation. Given the paucity of compounds that reduce hyperexcitability and neuron loss, have anti-inflammatory properties, and are well tolerated by the brain, D-serine, an endogenous amino acid, offers new hope as a therapeutic agent for refractory TLE.


Asunto(s)
Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/patología , Serina/uso terapéutico , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Conducta Animal , Encéfalo/patología , Corteza Entorrinal/efectos de los fármacos , Corteza Entorrinal/patología , Gliosis/patología , Inflamación/patología , Microglía/efectos de los fármacos , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Ratas Sprague-Dawley , Serina/administración & dosificación , Serina/farmacología
6.
Proc Natl Acad Sci U S A ; 117(45): 28402-28411, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33106420

RESUMEN

The circadian clock is based on a transcriptional feedback loop with an essential time delay before feedback inhibition. Previous work has shown that PERIOD (PER) proteins generate circadian time cues through rhythmic nuclear accumulation of the inhibitor complex and subsequent interaction with the activator complex in the feedback loop. Although this temporal manifestation of the feedback inhibition is the direct consequence of PER's cytoplasmic trafficking before nuclear entry, how this spatial regulation of the pacemaker affects circadian timing has been largely unexplored. Here we show that circadian rhythms, including wake-sleep cycles, are lengthened and severely unstable if the cytoplasmic trafficking of PER is disrupted by any disease condition that leads to increased congestion in the cytoplasm. Furthermore, we found that the time delay and robustness in the circadian clock are seamlessly generated by delayed and collective phosphorylation of PER molecules, followed by synchronous nuclear entry. These results provide clear mechanistic insight into why circadian and sleep disorders arise in such clinical conditions as metabolic and neurodegenerative diseases and aging, in which the cytoplasm is congested.


Asunto(s)
Citoplasma/metabolismo , Homeostasis , Transporte de Proteínas/fisiología , Sueño/fisiología , Células 3T3-L1 , Animales , Proteína 5 Relacionada con la Autofagia , Proteínas CLOCK/metabolismo , Línea Celular , Relojes Circadianos , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo
7.
IBRO Rep ; 9: 147-156, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32775760

RESUMEN

Glutamatergic N-methyl-d-aspartate receptors (NMDARs) are heterotetrameric proteins whose subunits are derived from three gene families, GRIN1 (codes for GluN1), GRIN2 (GluN2) and GRIN3 (GluN3). In addition to providing binding sites for glutamate and the co-agonist glycine, these subunits in their di (d-) and tri (t-) heteromeric configurations regulate various aspects of receptor function in the brain. For example, the decay kinetics of NMDAR-mediated synaptic currents depend on the type of GluN2 subunit (GluN2A-GluN2D) in the receptor subunit composition. While much is known about the contributions of GluN1 and GluN2 to d-NMDAR function, we know comparatively little about how GluN3 influences the function of t-NMDARs composed of one or more subunits from each of the three gene families. We report here that in addition to altering kinetics and voltage-dependent properties, the GluN3 subunit endows these receptors with ion selectivity wherein influx of Ca2+ is preferred over Na+. This became apparent in the process of assessing Ca2+ permeability through these receptors and is of significance given that NMDARs are generally believed to be nonselective to cations and increased selectivity can lead to enhanced permeability. This was true of two independent brain regions where t-NMDARs are expressed, the somatosensory cortex, where both receptor subtypes are expressed at separate inputs onto single neurons, and the entorhinal cortex, where they are co-expressed at individual synaptic inputs. Based on this data and the sequence of amino acids lining selectivity filters within these subunits, we propose GluN3 to be a regulatory subunit for ion selectivity in t-NMDARs.

8.
J Neurophysiol ; 121(1): 238-254, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30461362

RESUMEN

The subunit composition of N-methyl-d-aspartate receptors (NMDARs) at synaptic inputs onto a neuron can either vary or be uniform depending on the type of neuron and/or brain region. Excitatory pyramidal neurons in the frontal and somatosensory cortices (L5), for example, show pathway-specific differences in NMDAR subunit composition in contrast with the entorhinal cortex (L3), where we now show colocalization of NMDARs with distinct subunit compositions at individual synaptic inputs onto these neurons. Subunit composition was deduced electrophysiologically based on alterations of current-voltage relationship ( I-V) profiles, amplitudes, and decay kinetics of minimally evoked, pharmacologically isolated, NMDAR-mediated excitatory postsynaptic currents by known subunit-preferring antagonists. The I-Vs were outwardly rectifying in a majority of neurons assayed (~80%), indicating expression of GluN1/GluN2/GluN3-containing triheteromeric NMDARs ( t-NMDARs) and of the conventional type, reversing close to 0 mV with prominent regions of negative slope, in the rest of the neurons sampled (~20%), indicating expression of GluN1/GluN2-containing diheteromeric NMDARs ( d-NMDARs). Blocking t-NMDARs in neurons with outwardly rectifying I-Vs pharmacologically unmasked d-NMDARs, with all responses antagonized using D-AP5. Coimmunoprecipitation assays of membrane-bound protein complexes isolated from the medial entorhinal area using subunit-selective antibodies corroborated stoichiometry and together suggested the coexpression of t- and d-NMDARs at these synapses. Colocalization of functionally distinct NMDAR subtypes at individual synaptic inputs likely enhances the repertoire of pyramidal neurons for information processing and plasticity within the entorhinal cortex. NEW & NOTEWORTHY The subunit composition of a N-methyl-d-aspartate (NMDA) receptor, which dictates most aspects of its function, can vary between neurons in different brain regions and/or between synaptic inputs onto single neurons. Here we demonstrate colocalization of tri- and diheteromeric-NMDA receptors at the same/single synaptic input onto excitatory neurons in the entorhinal cortex. Synaptic colocalization of distinct NMDAR subtypes might endow entorhinal cortical neurons with the ability to encode distinct patterns of neuronal activity through single synapses.


Asunto(s)
Corteza Entorrinal/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Animales , Corteza Entorrinal/citología , Corteza Entorrinal/efectos de los fármacos , Expresión Génica , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Neurotransmisores/farmacología , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Sinapsis/efectos de los fármacos , Técnicas de Cultivo de Tejidos
9.
FASEB J ; 33(1): 126-139, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29965797

RESUMEN

The circadian clock is a critical regulator of immune function. We recently highlighted a role for the circadian clock in a mouse model of pulmonary inflammation. The epithelial clock protein Bmal1 was required to regulate neutrophil recruitment in response to inflammatory challenge. Bmal1 regulated glucocorticoid receptor (GR) recruitment to the neutrophil chemokine, CXC chemokine ligand 5 (CXCL5), providing a candidate mechanism. We now show that clock control of pulmonary neutrophilia persists without rhythmic glucocorticoid availability. Epithelial GR-null mice had elevated expression of proinflammatory chemokines in the lung under homeostatic conditions. However, deletion of GR in the bronchial epithelium blocked rhythmic CXCL5 production, identifying GR as required to confer circadian control to CXCL5. Surprisingly, rhythmic pulmonary neutrophilia persisted, despite nonrhythmic CXCL5 responses, indicating additional circadian control mechanisms. Deletion of GR in myeloid cells alone did not prevent circadian variation in pulmonary neutrophilia and showed reduced neutrophilic inflammation in response to dexamethasone treatment. These new data show GR is required to confer circadian control to some inflammatory chemokines, but that this alone is insufficient to prevent circadian control of neutrophilic inflammation in response to inhaled LPS, with additional control mechanisms arising in the myeloid cell lineage.-Ince, L. M., Zhang, Z., Beesley, S., Vonslow, R. M., Saer, B. R., Matthews, L. C., Begley, N., Gibbs, J. E., Ray, D. W., Loudon, A. S. I. Circadian variation in pulmonary inflammatory responses is independent of rhythmic glucocorticoid signaling in airway epithelial cells.


Asunto(s)
Ritmo Circadiano/inmunología , Células Epiteliales/inmunología , Macrófagos Peritoneales/inmunología , Neutrófilos/inmunología , Neumonía/inmunología , Receptores de Glucocorticoides/fisiología , Sistema Respiratorio/inmunología , Animales , Células Cultivadas , Quimiocina CXCL5/metabolismo , Ritmo Circadiano/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Glucocorticoides/farmacología , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Neumonía/patología , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Sistema Respiratorio/patología , Transducción de Señal
10.
Curr Biol ; 27(22): 3454-3467.e8, 2017 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-29103939

RESUMEN

Robustness in biology is the stability of phenotype under diverse genetic and/or environmental perturbations. The circadian clock has remarkable stability of period and phase that-unlike other biological oscillators-is maintained over a wide range of conditions. Here, we show that the high fidelity of the circadian system stems from robust degradation of the clock protein PERIOD. We show that PERIOD degradation is regulated by a balance between ubiquitination and deubiquitination, and that disruption of this balance can destabilize the clock. In mice with a loss-of-function mutation of the E3 ligase gene ß-Trcp2, the balance of PERIOD degradation is perturbed and the clock becomes dramatically unstable, presenting a unique behavioral phenotype unlike other circadian mutant animal models. We believe that our data provide a molecular explanation for how circadian phases, such as wake-sleep onset times, can become unstable in humans, and we present a unique mouse model to study human circadian disorders with unstable circadian rhythm phases.


Asunto(s)
Ritmo Circadiano/fisiología , Proteínas Circadianas Period/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo , Animales , Proteínas CLOCK/genética , Relojes Circadianos , Ritmo Circadiano/genética , Ratones , Modelos Animales , Proteínas Circadianas Period/fisiología , Proteolisis , Sueño/genética , Trastornos del Sueño del Ritmo Circadiano/genética , Trastornos del Sueño del Ritmo Circadiano/fisiopatología , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteínas con Repetición de beta-Transducina/genética
11.
J Alzheimers Dis ; 55(3): 1155-1166, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27814295

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the leading cause of dementia, and as its prevalence increases, so does its detrimental impact on society. The currently available therapies have limited efficacy, leaving AD patients on an irrevocably fatal path of this disease. OBJECTIVE: The purpose of this study was to test efficacy of a novel combinatorial treatment approach to alleviate AD-like pathology. METHODS: We selected four naturally occurring compounds and used them in different combinations to test their effect on AD-like pathology. Employing a well-established cell culture AD model system, we evaluated levels of several diverse biomarkers associated with a number of cellular pathways associated with AD. The readouts included: amyloid-ß peptides, anti-inflammatory and anti-apoptotic proteins, oxidative enzymes, and reactive oxygen species. RESULTS: Using this approach, we demonstrated that the compounds delivered in combination had higher efficacy than individual treatments. Specifically, we observed significant reduction in levels of the amyloid-ß peptides, as well as pro-inflammatory proteins and reactive oxygen species. Similarly, delivery of compounds in combination resulted in an increased expression of anti-apoptotic proteins and anti-oxidative enzymes. Collectively, these modifications in AD pathology biomarkers reflect a promising therapeutic and preventive strategy to combat this disease. CONCLUSION: The above findings support a novel therapeutic approach to address a currently unmet medical need, which would benefit not only AD patients and their caregivers, but also society as a whole.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Antipsicóticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Catequina/análogos & derivados , Catequina/farmacología , Línea Celular Tumoral , Citocinas/metabolismo , Combinación de Medicamentos , Humanos , Melatonina/farmacología , Ratones , Mutación/genética , Neuroblastoma/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Resveratrol , Estilbenos/farmacología , Transfección
12.
PLoS One ; 11(7): e0159618, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27459195

RESUMEN

Circadian clocks impact vital cardiac parameters such as blood pressure and heart rate, and adverse cardiac events such as myocardial infarction and sudden cardiac death. In mammals, the central circadian pacemaker, located in the suprachiasmatic nucleus of the hypothalamus, synchronizes cellular circadian clocks in the heart and many other tissues throughout the body. Cardiac ventricle explants maintain autonomous contractions and robust circadian oscillations of clock gene expression in culture. In the present study, we examined the relationship between intrinsic myocardial function and circadian rhythms in cultures from mouse heart. We cultured ventricular explants or dispersed cardiomyocytes from neonatal mice expressing a PER2::LUC bioluminescent reporter of circadian clock gene expression. We found that isoproterenol, a ß-adrenoceptor agonist known to increase heart rate and contractility, also amplifies PER2 circadian rhythms in ventricular explants. We found robust, cell-autonomous PER2 circadian rhythms in dispersed cardiomyocytes. Single-cell rhythms were initially synchronized in ventricular explants but desynchronized in dispersed cells. In addition, we developed a method for long-term, simultaneous monitoring of clock gene expression, contraction rate, and basal intracellular Ca2+ level in cardiomyocytes using PER2::LUC in combination with GCaMP3, a genetically encoded fluorescent Ca2+ reporter. In contrast to robust PER2 circadian rhythms in cardiomyocytes, we detected no rhythms in contraction rate and only weak rhythms in basal Ca2+ level. In summary, we found that PER2 circadian rhythms of cardiomyocytes are cell-autonomous, amplified by adrenergic signaling, and synchronized by intercellular communication in ventricle explants, but we detected no robust circadian rhythms in contraction rate or basal Ca2+.


Asunto(s)
Relojes Circadianos/fisiología , Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal , Animales , Calcio/metabolismo , Comunicación Celular , Células Cultivadas , Péptidos y Proteínas de Señalización del Ritmo Circadiano/genética , Péptidos y Proteínas de Señalización del Ritmo Circadiano/metabolismo , Regulación de la Expresión Génica , Ventrículos Cardíacos/efectos de los fármacos , Mediciones Luminiscentes , Ratones , Ratones Transgénicos , Contracción Miocárdica/genética , Proteínas Circadianas Period/genética , Función Ventricular/efectos de los fármacos , Función Ventricular/genética
13.
Neuropharmacology ; 101: 439-48, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26476274

RESUMEN

UNLABELLED: Bipolar disorder (BD) is associated with mood episodes and low amplitude circadian rhythms. Previously, we demonstrated that fibroblasts grown from BD patients show weaker amplification of circadian rhythms by lithium compared to control cells. Since calcium signals impact upon the circadian clock, and L-type calcium channels (LTCC) have emerged as genetic risk factors for BD, we examined whether loss of function in LTCCs accounts for the attenuated response to lithium in BD cells. We used fluorescent dyes to measure Ca(2+) changes in BD and control fibroblasts after lithium treatment, and bioluminescent reporters to measure Per2::luc rhythms in fibroblasts from BD patients, human controls, and mice while pharmacologically or genetically manipulating calcium channels. Longitudinal expression of LTCC genes (CACNA1C, CACNA1D and CACNB3) was then measured over 12-24 h in BD and control cells. Our results indicate that independently of LTCCs, lithium stimulated intracellular Ca(2+) less effectively in BD vs. control fibroblasts. In longitudinal studies, pharmacological inhibition of LTCCs or knockdown of CACNA1A, CACNA1C, CACNA1D and CACNB3 altered circadian rhythm amplitude. Diltiazem and knockdown of CACNA1C or CACNA1D eliminated lithium's ability to amplify rhythms. Knockdown of CACNA1A or CACNB3 altered baseline rhythms, but did not affect rhythm amplification by lithium. In human fibroblasts, CACNA1C genotype predicted the amplitude response to lithium, and the expression profiles of CACNA1C, CACNA1D and CACNB3 were altered in BD vs. CONTROLS: We conclude that in cells from BD patients, calcium signaling is abnormal, and that LTCCs underlie the failure of lithium to amplify circadian rhythms.


Asunto(s)
Canales de Calcio Tipo L/genética , Canales de Calcio/genética , Señalización del Calcio/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos , Ritmo Circadiano/genética , Litio/uso terapéutico , Adulto , Anciano , Animales , Trastorno Bipolar/genética , Trastorno Bipolar/patología , Compuestos de Boro/farmacología , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Femenino , Fibroblastos , Humanos , Litio/farmacología , Masculino , Ratones , Persona de Mediana Edad , Células 3T3 NIH , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Verapamilo/farmacología , Adulto Joven
14.
Nat Commun ; 6: 8587, 2015 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-26617050

RESUMEN

Self-sustaining oscillations are essential for diverse physiological functions such as the cell cycle, insulin secretion and circadian rhythms. Synthetic oscillators using biochemical feedback circuits have been generated in cell culture. These synthetic systems provide important insight into design principles for biological oscillators, but have limited similarity to physiological pathways. Here we report the generation of an artificial, mammalian circadian clock in vivo, capable of generating robust, tunable circadian rhythms. In mice deficient in Per1 and Per2 genes (thus lacking circadian rhythms), we artificially generate PER2 rhythms and restore circadian sleep/wake cycles with an inducible Per2 transgene. Our artificial clock is tunable as the period and phase of the rhythms can be modulated predictably. This feature, and other design principles of our work, might enhance the study and treatment of circadian dysfunction and broader aspects of physiology involving biological oscillators.


Asunto(s)
Trastornos Cronobiológicos/fisiopatología , Relojes Circadianos , Animales , Trastornos Cronobiológicos/genética , Trastornos Cronobiológicos/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Circadianas Period/deficiencia , Proteínas Circadianas Period/genética
15.
Mol Hum Reprod ; 21(8): 662-71, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25939854

RESUMEN

Circadian genes are expressed in virtually all cells and tissues, and circadian rhythms influence many bodily processes, including reproductive physiology. The expression of hMTNR1B is suppressed during pregnancy until late in term (much like the oxytocin receptor), at which time it is up-regulated to allow for the nocturnal melatonin/oxytocin synergy, which promotes strong nocturnal contractions. Little is currently known about the regulation of hMNTR1b, nor about its functional significance in the myometrium. We, therefore, aimed to elucidate some of the transcription factors that regulate hMNTR1b gene expression in the human myometrium and to determine if hMNTR1b is under circadian control. In this study, we used immortalized and primary myometrial cells that were assessed for circadian gene expression rhythms using real-time bioluminometry and quantitative PCR. Chromatin immunoprecipitation examined the binding of the clock gene product brain and muscle aryl hydrocarbon receptor nuclear translocator (ARNT)-like protein 1 (BMAL1) to the promoter of the hMTNR1B gene. Overexpression studies tested the role of circadian locomotor output cycles kaput (CLOCK) and its partner BMAL1 in regulating hMTNR1B expression. We confirmed circadian clock gene expression in both immortalized human myometrial cells and primary myometrial cell cultures. We further showed that the hBMAL1 protein binds to an E-box motif in the proximal promoter of the hMTNR1B gene. Overexpression studies demonstrated that the BMAL1/CLOCK complex activates expression of hMTNR1B leading to a circadian rhythm in phase with the E-box driven clock gene hPER2 (Period 2). These results indicate, for the first time, the presence of a functional circadian clock in the human myometrium with the hMTNR1B gene as a clock controlled target. Further investigations could open new vistas for understanding the regulation of uterine contractions and the timing of human labor.


Asunto(s)
Ritmo Circadiano/fisiología , Regulación de la Expresión Génica , Miocitos del Músculo Liso/metabolismo , Miometrio/metabolismo , Receptor de Melatonina MT2/genética , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Relojes Circadianos/genética , Relojes Circadianos/fisiología , Ritmo Circadiano/genética , Femenino , Humanos , Melatonina/metabolismo , Miometrio/citología , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Receptor de Melatonina MT2/metabolismo
16.
Fertil Steril ; 102(2): 329-35, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25015556

RESUMEN

OBJECTIVE: To review and update the research on melatonin receptor expression in the human myometrium, in particular as it pertains to uterine contractility at labor. DESIGN: Summary of previous studies with the addition of new data on the transcriptional regulation of melatonin receptor expression in human myometrial cells. SETTING: Not applicable. PATIENT(S): Late-term pregnant volunteers. INTERVENTION(S): Biopsy collection for in vitro analyses provided the original data. More recently, uterine contractions in late-term pregnant volunteers were assessed before, during, and after acute white-light exposure. MAIN OUTCOME MEASURE(S): Melatonin receptor signaling in myometrial cells and uterine contractions in late-term pregnant volunteers. RESULT(S): Melatonin acts through the MTNR1B melatonin receptor that is expressed in the myometrium at late term to synergistically enhance oxytocin-dependent signaling and contractions. Acute inhibition of endogenous melatonin levels with light reversibly suppresses uterine contractions. CONCLUSION(S): These results point to a significant role for circulating melatonin in the timing and degree of uterine contractions in late-term pregnancy. Understanding the regulation of melatonin receptors remains a future objective.


Asunto(s)
Melatonina/metabolismo , Miometrio/metabolismo , Receptores de Melatonina/metabolismo , Contracción Uterina , Relojes Circadianos , Ritmo Circadiano , Femenino , Humanos , Luz , Miometrio/efectos de la radiación , Oxitocina/metabolismo , Fotoperiodo , Embarazo , Receptores de Melatonina/efectos de la radiación , Transducción de Señal , Factores de Tiempo , Contracción Uterina/efectos de la radiación
17.
PLoS One ; 7(3): e33292, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22428012

RESUMEN

Lithium salt has been widely used in treatment of Bipolar Disorder, a mental disturbance associated with circadian rhythm disruptions. Lithium mildly but consistently lengthens circadian period of behavioural rhythms in multiple organisms. To systematically address the impacts of lithium on circadian pacemaking and the underlying mechanisms, we measured locomotor activity in mice in vivo following chronic lithium treatment, and also tracked clock protein dynamics (PER2::Luciferase) in vitro in lithium-treated tissue slices/cells. Lithium lengthens period of both the locomotor activity rhythms, as well as the molecular oscillations in the suprachiasmatic nucleus, lung tissues and fibroblast cells. In addition, we also identified significantly elevated PER2::LUC expression and oscillation amplitude in both central and peripheral pacemakers. Elevation of PER2::LUC by lithium was not associated with changes in protein stabilities of PER2, but instead with increased transcription of Per2 gene. Although lithium and GSK3 inhibition showed opposing effects on clock period, they acted in a similar fashion to up-regulate PER2 expression and oscillation amplitude. Collectively, our data have identified a novel amplitude-enhancing effect of lithium on the PER2 protein rhythms in the central and peripheral circadian clockwork, which may involve a GSK3-mediated signalling pathway. These findings may advance our understanding of the therapeutic actions of lithium in Bipolar Disorder or other psychiatric diseases that involve circadian rhythm disruptions.


Asunto(s)
Conducta Animal/efectos de los fármacos , Relojes Circadianos/efectos de los fármacos , Litio/farmacología , Locomoción/efectos de los fármacos , Proteínas Circadianas Period/metabolismo , Análisis de Varianza , Animales , Conducta Animal/fisiología , Relojes Circadianos/fisiología , Cartilla de ADN/genética , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Locomoción/fisiología , Luciferasas/metabolismo , Mediciones Luminiscentes , Pulmón/efectos de los fármacos , Pulmón/fisiología , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Núcleo Supraquiasmático/efectos de los fármacos , Núcleo Supraquiasmático/fisiología , Factores de Tiempo
18.
Proc Natl Acad Sci U S A ; 109(2): 582-7, 2012 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22184247

RESUMEN

Diurnal variation in inflammatory and immune function is evident in the physiology and pathology of humans and animals, but molecular mechanisms and mediating cell types that provide this gating remain unknown. By screening cytokine responses in mice to endotoxin challenge at different times of day, we reveal that the magnitude of response exhibited pronounced temporal dependence, yet only within a subset of proinflammatory cytokines. Disruption of the circadian clockwork in macrophages (primary effector cells of the innate immune system) by conditional targeting of a key clock gene (bmal1) removed all temporal gating of endotoxin-induced cytokine response in cultured cells and in vivo. Loss of circadian gating was coincident with suppressed rev-erbα expression, implicating this nuclear receptor as a potential link between the clock and inflammatory pathways. This finding was confirmed in vivo and in vitro through genetic and pharmacological modulation of REV-ERBα activity. Circadian gating of endotoxin response was lost in rev-erbα(-/-) mice and in cultured macrophages from these animals, despite maintenance of circadian rhythmicity within these cells. Using human macrophages, which show circadian clock gene oscillations and rhythmic endotoxin responses, we demonstrate that administration of a synthetic REV-ERB ligand, or genetic knockdown of rev-erbα expression, is effective at modulating the production and release of the proinflammatory cytokine IL-6. This work demonstrates that the macrophage clockwork provides temporal gating of systemic responses to endotoxin, and identifies REV-ERBα as the key link between the clock and immune function. REV-ERBα may therefore represent a unique therapeutic target in human inflammatory disease.


Asunto(s)
Ritmo Circadiano/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/inmunología , Interleucina-6/inmunología , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/inmunología , Factores de Transcripción ARNTL/genética , Análisis de Varianza , Animales , Endotoxinas/toxicidad , Humanos , Macrófagos/inmunología , Ratones , Ratones Noqueados , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Factores de Tiempo
19.
PLoS One ; 6(1): e15766, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21267416

RESUMEN

CONTEXT: Cortisol is released in ultradian pulses. The biological relevance of the resulting fluctuating cortisol concentration has not been explored. OBJECTIVE: Determination of the biological consequences of ultradian cortisol pulsatility. DESIGN: A novel flow through cell culture system was developed to deliver ultradian pulsed or continuous cortisol to cells. The effects of cortisol dynamics on cell proliferation and survival, and on gene expression were determined. In addition, effects on glucocorticoid receptor (GR) expression levels and phosphorylation, as a potential mediator, were measured. RESULTS: Pulsatile cortisol caused a significant reduction in cell survival compared to continuous exposure of the same cumulative dose, due to increased apoptosis. Comprehensive analysis of the transcriptome response by microarray identified genes with a differential response to pulsatile versus continuous glucocorticoid delivery. These were confirmed with qRT-PCR. Several transcription factor binding sites were enriched in these differentially regulated target genes, including CCAAT-displacement protein (CDP). A CDP regulated reporter gene (MMTV-luc) was, as predicted, also differentially regulated by pulsatile compared to continuous cortisol delivery. Importantly there was no effect of cortisol delivery kinetics on either GR expression, or activation (GR phosphoSer(211)). CONCLUSIONS: Cortisol oscillations exert important effects on target cell gene expression, and phenotype. This is not due to differences in cumulative cortisol exposure, or either expression, or activation of the GR. This suggests a novel means to regulate GR function.


Asunto(s)
Ciclos de Actividad/fisiología , Proliferación Celular , Supervivencia Celular , Perfilación de la Expresión Génica , Hidrocortisona/fisiología , Técnicas de Cultivo de Célula/métodos , Células HeLa , Humanos , Fosforilación , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transducción de Señal
20.
J Cell Sci ; 121(Pt 21): 3629-35, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18946026

RESUMEN

The nuclear receptor REV-ERBalpha is a key negative-feedback regulator of the biological clock. REV-ERBalpha binds to ROR elements of the Bmal1 (Arntl) promoter and represses Bmal1 transcription. This stabilizing negative loop is important for precise control of the circadian pacemaker. In the present study, we identified a novel synthetic REV-ERBalpha ligand, which enhances the recruitment of nuclear receptor co-repressor (NCoR) to REV-ERBalpha. In order to explore REV-ERBalpha action on resetting responses of the molecular clock, we first established the rhythmic transcription profile and expression level of REV-ERBalpha in Rat-1 fibroblasts. When applied at different phases of the circadian oscillation to cell models containing stably transfected Bmal1::Luc or Per2::Luc, the REV-ERBalpha ligand induced phase-dependent bi-directional phase shifts. When the phase changes were plotted against time, a clear phase response curve was revealed, with a significant peak-to-trough amplitude of ca. 5 hours. The phase-resetting effect was also observed when the compound was applied to primary lung fibroblasts and ectopic lung slices from transgenic PER2::Luc mice. Therefore, similar regulation of REV-ERBalpha function by endogenous ligands, such as heme, is likely to be an important mechanism for clock resetting. In addition, we identify a new means to generate phasic shifts in the clock.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Relojes Biológicos , Ritmo Circadiano , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Genes Reporteros , Ligandos , Luminiscencia , Pulmón/citología , Pulmón/metabolismo , Ratones , Modelos Biológicos , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares , Oscilometría , Ratas , Receptores Citoplasmáticos y Nucleares/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...