Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Vis Exp ; (157)2020 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-32202523

RESUMEN

Profound transcriptional heterogeneity in anatomically adjacent single cells suggests that robust tissue functionality may be achieved by cellular phenotype diversity. Single-cell experiments investigating the network dynamics of biological systems demonstrate cellular and tissue responses to various conditions at biologically meaningful resolution. Herein, we explain our methods for gathering single cells from anatomically specific locations and accurately measuring a subset of their gene expression profiles. We combine laser capture microdissection (LCM) with microfluidic reverse transcription quantitative polymerase chain reactions (RT-qPCR). We also use this microfluidic RT-qPCR platform to measure the microbial abundance of gut contents.


Asunto(s)
Captura por Microdisección con Láser/métodos , Microfluídica , Trastornos Relacionados con Opioides/diagnóstico , Trastornos Relacionados con Opioides/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de la Célula Individual , Biología de Sistemas , Transcriptoma , Animales , Deshidratación , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Microbioma Gastrointestinal/genética , Redes Reguladoras de Genes , Masculino , Ratas Sprague-Dawley , Transcripción Genética
2.
Cells ; 8(9)2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31438578

RESUMEN

BKCa channels, originally discovered in Drosophila melanogaster as slowpoke (slo), are recognized for their roles in cellular and organ physiology. Pharmacological approaches implicated BKCa channels in cellular and organ protection possibly for their ability to modulate mitochondrial function. However, the direct role of BKCa channels in regulating mitochondrial structure and function is not deciphered. Here, we demonstrate that BKCa channels are present in fly mitochondria, and slo mutants show structural and functional defects in mitochondria. slo mutants display an increase in reactive oxygen species and the modulation of ROS affected their survival. We also found that the absence of BKCa channels reduced the lifespan of Drosophila, and overexpression of human BKCa channels in flies extends life span in males. Our study establishes the presence of BKCa channels in mitochondria of Drosophila and ascertains its novel physiological role in regulating mitochondrial structural and functional integrity, and lifespan.


Asunto(s)
Drosophila melanogaster/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Longevidad , Mitocondrias/metabolismo , Animales
3.
Front Neurosci ; 13: 665, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31333398

RESUMEN

Drug-seeking in opioid dependence is due in part to the severe negative emotion associated with the withdrawal syndrome. It is well-established that negative emotional states emerge from activity in the amygdala. More recently, gut microflora have been shown to contribute substantially to such emotions. We measured gene expression in single glia and neurons gathered from the amygdala using laser capture microdissection and simultaneously measured gut microflora in morphine-dependent and withdrawn rats to investigate drivers of negative emotion in opioid withdrawal. We found that neuroinflammatory genes, notably Tnf, were upregulated in the withdrawal condition and that astrocytes, in particular, were highly active. We also observe a decreased Firmicutes to Bacteroides ratio in opioid withdrawal indicating gut dysbiosis. We speculate that these inflammatory and gut microflora changes contribute to the negative emotion experienced in opioid withdrawal that motivates dependence.

4.
Neurobiol Stress ; 11: 100176, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31236436

RESUMEN

The endocannabinoid (eCB) system has been implicated in a variety of physiological functions due to abundant expression of its receptors and endogenous ligands in the central nervous system. Substantial progress has been made in understanding how the eCB system influences the brain norepinephrine (NE) system, an important neurochemical target in the continued development of new therapies for stress-induced psychiatric disorders. We, and others, have characterized the neuroanatomical, biochemical and pharmacological effects of cannabinoid receptor modulation on brain noradrenergic circuitry and defined how molecular elements of the eCB system are positioned to directly impact the locus coeruleus (LC)-prefrontal cortex pathway, a neural circuit well recognized for contributing to symptoms of hyperarousal, a key pathophysiological feature of stress-related disorders. We also described molecular and electrophysiological properties of LC noradrenergic neurons and NE release in the medial prefrontal cortex under conditions of cannabinoid type 1 receptor deletion. Finally, we identified how stress influences cannabinoid modulation of the coeruleo-cortical pathway. A number of significant findings emerged from these studies that will be summarized in the present review and have important implications for clinical studies targeting the eCB system in the treatment of stress-induced psychiatric disorders.

5.
Brain Struct Funct ; 224(7): 2385-2405, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31250157

RESUMEN

A culmination of evidence from the literature points to the Locus Coeruleus (LC)-Norepinephrine system as an underappreciated and understudied area of research in the context of Alzheimer's Disease (AD). Stress is a risk factor for developing AD, and is supported by multiple clinical and preclinical studies demonstrating that amplification of the stress system disrupts cellular and molecular processes at the synapse, promoting the production and accumulation of the amyloid beta (Aß42) peptide. Stress-induced activation of the LC is mediated by corticotropin releasing factor (CRF) and CRF receptors exhibit sex-biased stress signaling. Sex differences are evident in the neurochemical, morphological and molecular regulation of LC neurons by CRF, providing a compelling basis for the higher prevalence of stress-related disorders such as AD in females. In the present study, we examined the cellular substrates for interactions between Aß and tyrosine hydroxylase a marker of noradrenergic somatodendritic processes in the LC, and Dopamine-ß-Hydroxylase (DßH) in the infralimbic medial prefrontal cortex (ILmPFC) in mice conditionally overexpressing CRF in the forebrain (CRFOE) under a Doxycycline (DOX) regulated tetO promoter. CRFOE was sufficient to elicit a redistribution of Aß peptides in the somatodendritic processes of the LC of male and female transgenic mice, without altering total Aß42 protein expression levels. DOX treated groups exhibited lysosomal compartments with apparent lipofuscin and abnormal morphology, indicating potential dysfunction of these Aß42-clearing compartments. In female DOX treated groups, swollen microvessels with lipid-laden vacuoles were also observed, a sign of blood-brain-barrier dysfunction. Finally, sex differences were observed in the prefrontal cortex, as females responded to DOX treatment with increased frequency of co-localization of Aß42 and DßH in noradrenergic axon terminals compared to vehicle treated controls, while male groups showed no significant changes. We hypothesize that the observed sex differences in Aß42 distribution in this model of CRF hypersignaling is based on increased responsivity of female rodent CRFR1 in the LC. Aß42 production is enhanced during increased neuronal activation, therefore, the excitation of DOX treated female CRFOE LC neurons projecting to the mPFC may exhibit more frequent co-localization with Aß due to increased neuronal activity of noradrenergic neurons.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Péptidos beta-Amiloides/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Locus Coeruleus/metabolismo , Animales , Femenino , Masculino , Ratones , Norepinefrina/metabolismo , Corteza Prefrontal/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo
6.
Eur Neuropsychopharmacol ; 29(4): 482-492, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30878321

RESUMEN

PTSD is heterogeneous disorder that can be long lasting and often has delayed onset following exposure to a traumatic event. Therefore, it is important to take a staging approach to evaluate progression of biological mechanisms of the disease. Here, we begin to evaluate the temporal trajectory of changes following exposure to traumatic stressors in the SPS rat PTSD model. The percent of animals displaying severe anxiety on EPM increased from 17.5% at one week to 57.1% two weeks after SPS stressors, indicating delayed onset or progressive worsening of the symptoms. The LC displayed prolonged activation, and dysbalance of the CRH/NPY systems, with enhanced CRHR1 gene expression, coupled with reduced mRNAs for NPY and Y2R. In the mediobasal hypothalamus, increased CRH mRNA levels were sustained, but there was a flip in alterations of HPA regulatory molecules, GR and FKBP5 and Y5 receptor at two weeks compared to one week. Two weeks after SPS, intranasal NPY at 300 µg/rat, but not 150 µg which was effective after one week, reversed SPS triggered elevated anxiety. It also reversed SPS elicited depressive/despair symptoms and hyperarousal. Overall, the results reveal time-dependent progression in development of anxiety symptoms and molecular impairments in gene expression for CRH and NPY systems in LC and mediobasal hypothalamus by SPS. With longer time afterwards only a higher dose of NPY was effective in reversing behavioral impairments triggered by SPS, indicating that therapeutic approaches should be adjusted according to the degree of biological progression of the disorder.


Asunto(s)
Expresión Génica , Hipotálamo/metabolismo , Locus Coeruleus/metabolismo , Neuropéptido Y/farmacología , Trastornos por Estrés Postraumático/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Hormona Liberadora de Corticotropina/biosíntesis , Masculino , Neuropéptido Y/biosíntesis , Ratas , Receptores de Hormona Liberadora de Corticotropina/biosíntesis , Receptores de Neuropéptido Y/biosíntesis , Proteínas de Unión a Tacrolimus/biosíntesis , Factores de Tiempo
7.
Brain Struct Funct ; 224(4): 1429-1446, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30767070

RESUMEN

Stress-related psychiatric diseases are nearly twice as prevalent in women compared to men. We recently showed in male rats that the resident-intruder model of social stress differentially engages stress-related circuitry that regulates norepinephrine-containing neurons of the locus coeruleus (LC) depending on coping strategy as determined by the latency to assume a defeat posture. Here, we determined whether this social stress had similar effects in female rats. LC afferents were retrogradely labeled with Fluorogold (FG) and rats had one or five daily exposures to an aggressive resident. Sections through the nucleus paragigantocellularis (PGi), a source of enkephalin (ENK) afferents to the LC, and central nucleus of the amygdala (CeA), a source of corticotropin-releasing factor (CRF) afferents to the LC, were processed for immunocytochemical detection of c-fos, a marker of neuronal activity, FG and ENK or CRF. Like male rats, female rats defeated with a relatively short latency (SL) in response to a single resident-intruder exposure and showed significant c-fos activation of LC neurons, PGi-ENK LC afferents, and CeA-CRF-LC afferents. With repeated exposure, some rats exhibited a long latency to defeat (LL). LC neurons and CeA-CRF-LC afferents were activated in SL rats compared to control and LL, whereas PGi-ENK LC afferents were not. Conversely, in LL rats, PGi-ENK LC and CeA-CRF-LC afferents were activated compared to controls but not LC neurons. CRF type 1 receptor (CRF1) and µ-opioid receptor (MOR) expression levels in LC were decreased in LL rats. Finally, electron microscopy showed a relative increase in MOR on the plasma membrane of LL rats and a relative increase in CRF1 on the plasma membrane of SL rats. Together, these results suggest that as is the case for males, social stress engages divergent circuitry to regulate the LC in female rats depending on coping strategy, with a bias towards CRF influence in more subordinate rats and opioid influence in less subordinate rats.


Asunto(s)
Adaptación Psicológica/fisiología , Locus Coeruleus/metabolismo , Conducta Social , Estrés Psicológico/metabolismo , Animales , Núcleo Amigdalino Central/metabolismo , Femenino , Locus Coeruleus/ultraestructura , Bulbo Raquídeo/metabolismo , Vías Nerviosas/metabolismo , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores Opioides mu/metabolismo
8.
Brain Res ; 1702: 46-53, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29577889

RESUMEN

The evolution of peptidergic signaling systems in the central nervous system serves a distinct and crucial role in brain processes and function. The diversity of physiological peptides and the complexity of their regulation and secretion from the dense core vesicles (DCV) throughout the brain is a topic greatly in need of investigation, though recent years have shed light on cellular and molecular mechanisms that are summarized in this review. Here, we focus on the convergence of peptidergic systems onto the Locus Coeruleus (LC), the sole provider of norepinephrine (NE) to the cortex and hippocampus, via large DCV. As the LC-NE system is one of the first regions of the brain to undergo degeneration in Alzheimer's Disease (AD), and markers of DCV have consistently been demonstrated to have biomarker potential for AD progression, here we summarize the current literature linking the LC-NE system with DCV dysregulation and Aß peptides. We also include neuroanatomical data suggesting that the building blocks of senile plaques, Aß monomers, may be localized to DCV of the LC and noradrenergic axon terminals of the prefrontal cortex. Finally, we explore the putative consequences of chronic stress on Aß production and the role that DCV may play in LC degeneration. Clinical data of immunological markers of DCV in AD patients are discussed.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Locus Coeruleus/fisiología , Vesículas Secretoras/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Neuronas/metabolismo , Neuropéptidos/metabolismo , Norepinefrina/metabolismo , Norepinefrina/fisiología , Vesículas Secretoras/patología
9.
Brain Res ; 1706: 75-85, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30391476

RESUMEN

The locus coeruleus (LC)-norepinephrine (NE) system is a key nucleus in which endogenous opioid and stress systems intersect to regulate the stress response. LC neurons of male rats become sensitized to stress following chronic morphine administration. Whether sex dictates this pattern of opioid-induced plasticity has not been demonstrated. Delineating the neurobiological adaptations produced by chronic opioids will enhance our understanding of stress vulnerability in opioid-dependent individuals, and may reveal how stress negatively impacts addiction recovery. In the present study, the effect of chronic morphine on the subcellular distribution of mu-opioid (MOR) and CRF receptors (CRFR) was investigated in the LC of male and female rats using immunoelectron microscopy. Results showed that placebo-treated females exhibited higher MOR and CRFR cytoplasmic distribution ratio when compared to placebo-treated males. Chronic morphine exposure induced a shift in the distribution of MOR immunogold-silver particles from the plasma membrane to the cytoplasm selectively in male LC neurons. Interestingly, chronic morphine exposure induced CRFR recruitment to the plasma membrane of both male and female LC neurons. These findings provide a potential mechanism by which chronic opioid administration increases stress vulnerability in males and females via an increase in surface availability of CRFR in LC neurons. However, our results also support the notion that cellular adaptations to chronic opioids differ across the sexes as redistribution of MOR following morphine exposure was only observed in male LC neurons.


Asunto(s)
Locus Coeruleus/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Receptores Opioides mu/efectos de los fármacos , Analgésicos Opioides/farmacología , Animales , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Masculino , Microscopía Inmunoelectrónica/métodos , Morfina/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Norepinefrina/metabolismo , Péptidos Opioides/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores Opioides mu/metabolismo , Rombencéfalo/efectos de los fármacos , Caracteres Sexuales , Factores Sexuales , Estrés Fisiológico/fisiología
10.
Eur J Neurosci ; 48(5): 2118-2138, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30103253

RESUMEN

Cannabinoids are capable of modulating mood, arousal, cognition and behavior, in part via their effects on the noradrenergic nucleus locus coeruleus (LC). Dysregulation of LC signaling and norepinephrine (NE) efflux in the medial prefrontal cortex (mPFC) can lead to the development of psychiatric disorders, and CB1r deletion results in alterations of α2- and ß1-adrenoceptors in the mPFC, suggestive of increased LC activity. To determine how CB1r deletion alters LC signaling, whole-cell patch-clamp electrophysiology was conducted in LC-NE neurons of male and female wild type (WT) and CB1r-knock out (KO) mice. CB1r deletion caused a significant increase in LC-NE excitability and input resistance in male but not female mice when compared to WT. CB1r deletion also caused adaptations in several indices of noradrenergic function. CB1r/CB2r-KO male mice had a significant increase in cortical NE levels and tyrosine hydroxylase and CRF levels in the LC compared to WT males. CB1r/CB2r-KO female mice showed a significant increase in LC α2-AR levels compared to WT females. To further probe actions of the endocannabinoid system as an anti-stress neuromediator, the effect of CB1r deletion on CRF-induced responses in the LC was investigated. The increase in LC-NE excitability observed in male and female WT mice following CRF (300 nM) bath application was not observed in CB1r-KO mice. These results indicate that cellular adaptations following CB1r deletion cause a disruption in LC-NE signaling in males but not females, suggesting underlying sex differences in compensatory mechanisms in KO mice as well as basal endocannabinoid regulation of LC-NE activity.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Norepinefrina/farmacología , Receptor Cannabinoide CB1/metabolismo , Animales , Cannabinoides/metabolismo , Femenino , Locus Coeruleus/efectos de los fármacos , Masculino , Ratones Noqueados , Neuronas/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptor Cannabinoide CB1/deficiencia , Caracteres Sexuales , Tirosina 3-Monooxigenasa/metabolismo
11.
Neuroscience ; 384: 21-40, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29775703

RESUMEN

As a neurochemical mediator of stress resilience, NPY has been shown to oppose excitatory effects of the pro-stress neuropeptide corticotropin-releasing factor (CRF). Previous studies have described the anatomical organization of NPY and CRF in the central nucleus of the amygdala, which sends CRF projections to the locus coeruleus (LC), activating LC norepinephrine release. However, the cellular substrates for interactions between NPY and CRF in the LC remain unknown. In this study, we investigated these anatomical substrates in the male rat LC, using immunocytochemistry, confocal microscopy, and immunoelectron microscopy to detect NPY and CRF, as well as CRF and Y1 or Y2 receptors (Y1R or Y2R). Immunofluorescence and electron microscopy revealed both co-localization of NPY and CRF in LC axon terminals, as well as separately labeled terminals, suggesting NPY and CRF may serve as co-transmitters in a subset of terminals. Semi-quantitative analysis showed that 32.4% of CRF-labeled terminals contained NPY, while 58.2% (152/261) of NPY-labeled terminals contained CRF. With respect to Y1R and CRF, dual immunoelectron microscopy showed that 23.3% (67/288) of CRF-labeled axon terminals directly contacted Y1R-labeled dendrites, while only 6.3% (18/288) of CRF-labeled axon terminals co-localized with Y1R. Dual immunoelectron microscopy also showed Y2R co-localized with 30.4% (103/339) CRF-labeled terminals, but only with 16.2% (55/339) of dendrites post-synaptic to CRF-labeled axon terminals in the LC. Taken together, these findings indicate multiple sites of interaction between CRF and NPY in the LC and suggest that conditions or drugs that modulate the NPY:CRF balance in the LC may promote stress resilience.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Locus Coeruleus/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Animales , Locus Coeruleus/ultraestructura , Masculino , Microscopía Inmunoelectrónica , Neuronas/ultraestructura , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Receptores de Neuropéptido Y/metabolismo
12.
Brain Struct Funct ; 223(1): 267-284, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28779307

RESUMEN

The locus coeruleus (LC)-norepinephrine (NE) system is an understudied circuit in the context of Alzheimer's disease (AD), and is thought to play an important role in neurodegenerative and neuropsychiatric diseases involving catecholamine neurotransmitters. Understanding the expression and distribution of the amyloid beta (Aß) peptide, a primary component of AD, under basal conditions and under conditions of NE perturbation within the coeruleo-cortical pathway may be important for understanding its putative role in pathological states. Thus, the goal of this study is to define expression levels and the subcellular distribution of endogenous Aß with respect to noradrenergic profiles in the rodent LC and medial prefrontal cortex (mPFC) and, further, to determine the functional relevance of NE in modulating endogenous Aß42 levels. We report that endogenous Aß42 is localized to tyrosine hydroxylase (TH) immunoreactive somatodendritic profiles of the LC and dopamine-ß-hydroxylase (DßH) immunoreactive axon terminals of the infralimbic mPFC (ILmPFC). Male and female naïve rats have similar levels of amyloid precursor protein (APP) cleavage products demonstrated by western blot, as well as similar levels of endogenous Aß42 as determined by enzyme-linked immunosorbent assay. Two models of NE depletion, DSP-4 lesion and DßH knockout (KO) mice, were used to assess the functional relevance of NE on endogenous Aß42 levels. DSP-4 lesioned rats and DßH-KO mice show significantly lower levels of endogenous Aß42. Noradrenergic depletion did not change APP-cleavage products resulting from ß-secretase processing. Thus, resultant decreases in endogenous Aß42 may be due to decreased neuronal activity of noradrenergic neurons, or, by decreased stimulation of adrenergic receptors which are known to contribute to Aß42 production by enhancing γ-secretase processing under normal physiological conditions.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Corteza Cerebral/metabolismo , Locus Coeruleus/metabolismo , Norepinefrina/deficiencia , Fragmentos de Péptidos/metabolismo , Proteína ADAM10/metabolismo , Neuronas Adrenérgicas/efectos de los fármacos , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/ultraestructura , Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Bencilaminas/toxicidad , Corteza Cerebral/ultraestructura , Dopamina beta-Hidroxilasa/deficiencia , Dopamina beta-Hidroxilasa/genética , Dopamina beta-Hidroxilasa/ultraestructura , Femenino , Locus Coeruleus/ultraestructura , Masculino , Ratones Noqueados , Microscopía Electrónica , Vías Nerviosas/metabolismo , Inhibidores de la Captación de Neurotransmisores/toxicidad , Fragmentos de Péptidos/ultraestructura , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/ultraestructura , Tirosina 3-Monooxigenasa/metabolismo
13.
Kidney Int ; 92(5): 1100-1118, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28754555

RESUMEN

Elastin deficiency causes vascular stiffening, a leading risk for hypertension and chronic kidney disease (CKD). The mechanisms mediating hypertension and/or CKD pathogenesis due to elastin deficiency are poorly understood. Using the elastin heterozygous (Eln+/-) mouse model, we tested whether renal dysfunction due to elastin deficiency occurs independently of and precedes the development of hypertension. We assessed blood pressure and renal hemodynamics in 30-day and 12-week-old male and female mice. At P30, blood pressure of Eln+/- mice was similar to wild-type controls; however, renal blood flow was lower, whereas renal vascular resistance was augmented at baseline in Eln+/- mice. At 12 weeks, renal vascular resistance remained elevated while filtration fraction was higher in male Eln+/- relative to wild-type mice. Heterozygous mice showed isolated systolic hypertension that was evident only at nighttime. Acute salt loading with 6% dietary sodium increased daytime systolic blood pressure only in male Eln+/- mice, causing a rightward shift and blunted slope of the pressure-natriuresis curve. Renal interlobar artery basal tone and myogenic response to increasing intraluminal pressure at day 10 were similar, whereas they were augmented at day 30 and at 12 weeks old in Eln+/- mice, and normalized by the AT1R blocker, candesartan. Heterozygous mice also exhibited podocyte foot process damage that persisted even when blood pressure was normalized to wild-type levels with hydralazine. Thus, elastin insufficiency triggers structural defects and abnormal remodeling of renal vascular signaling involving AT1R-mediated vascular mechanotransduction and renal hyperfiltration with increased blood pressure sensitivity to dietary sodium contributing to systolic hypertension.


Asunto(s)
Elastina/deficiencia , Hipertensión/etiología , Riñón/irrigación sanguínea , Insuficiencia Renal Crónica/etiología , Resistencia Vascular , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Bencimidazoles/farmacología , Compuestos de Bifenilo , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Elastina/genética , Femenino , Humanos , Riñón/metabolismo , Riñón/patología , Masculino , Mecanotransducción Celular/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor de Angiotensina Tipo 1/metabolismo , Eliminación Renal , Insuficiencia Renal Crónica/patología , Transducción de Señal , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio Dietético/metabolismo , Cloruro de Sodio Dietético/orina , Tetrazoles/farmacología
14.
Brain Struct Funct ; 222(7): 3007-3023, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28255675

RESUMEN

The noradrenergic system has been shown to play a key role in the regulation of stress responses, arousal, mood, and emotional states. Corticotropin-releasing factor (CRF) is a primary mediator of stress-induced activation of noradrenergic neurons in the nucleus locus coeruleus (LC). The endocannabinoid (eCB) system also plays a key role in modulating stress responses, acting as an "anti-stress" neuro-mediator. In the present study, we investigated the cellular sites for interactions between the cannabinoid receptor type 1 (CB1r) and CRF in the LC. Immunofluorescence and high-resolution immunoelectron microscopy showed co-localization of CB1r and CRF in both the core and peri-LC areas. Semi-quantitative analysis revealed that 44% (208/468) of CRF-containing axon terminals in the core and 35% (104/294) in the peri-LC expressed CB1r, while 18% (85/468) of CRF-containing axon terminals in the core and 6.5% (19/294) in the peri-LC were presynaptic to CB1r-containing dendrites. In the LC core, CB1r + CRF axon terminals were more frequently of the symmetric (inhibitory) type; while in the peri-LC, a majority were of the asymmetric (excitatory) type. Triple label immunofluorescence results supported the ultrastructural analysis indicating that CB1r + CRF axon terminals contained either gamma amino butyric acid or glutamate. Finally, anterograde transport from the central nucleus of the amygdala revealed that CRF-amygdalar afferents projecting to the LC contain CB1r. Taken together, these results indicate that the eCB system is poised to directly modulate stress-integrative heterogeneous CRF afferents in the LC, some of which arise from limbic sources.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Vías Aferentes/fisiología , Hormona Liberadora de Corticotropina/metabolismo , Locus Coeruleus/citología , Receptor Cannabinoide CB1/metabolismo , Neuronas Adrenérgicas/ultraestructura , Animales , Ácido Glutámico/metabolismo , Locus Coeruleus/metabolismo , Locus Coeruleus/ultraestructura , Masculino , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/ultraestructura , Tinción con Nitrato de Plata , Sinapsis/metabolismo , Sinapsis/ultraestructura , Sinaptofisina/metabolismo , Ácido gamma-Aminobutírico/metabolismo
15.
Brain Struct Funct ; 222(2): 1007-1026, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27376372

RESUMEN

It is well established that central nervous system norepinephrine (NE) and corticotropin-releasing factor (CRF) systems are important mediators of behavioral responses to stressors. More recent studies have defined a role for delta opioid receptors (DOPR) in maintaining emotional valence including anxiety. The amygdala plays an important role in processing emotional stimuli, and has been implicated in the development of anxiety disorders. Activation of DOPR or inhibition of CRF in the amygdala reduces baseline and stress-induced anxiety-like responses. It is not known whether CRF- and DOPR-containing amygdalar neurons interact or whether they are regulated by NE afferents. Therefore, this study sought to better define interactions between the CRF, DOPR and NE systems in the basolateral (BLA) and central nucleus of the amygdala (CeA) of the male rat using anatomical and functional approaches. Irrespective of the amygdalar subregion, dual immunofluorescence microscopy showed that DOPR was present in CRF-containing neurons. Immunoelectron microscopy confirmed that DOPR was localized to both dendritic processes and axon terminals in the BLA and CeA. Semi-quantitative dual immunoelectron microscopy analysis of gold-silver labeling for DOPR and immunoperoxidase labeling for CRF revealed that 55 % of the CRF neurons analyzed contained DOPR in the BLA while 67 % of the CRF neurons analyzed contained DOPR in the CeA. Furthermore, approximately 41 % of DOPR-labeled axon terminals targeted BLA neurons that expressed CRF while 29 % of DOPR-labeled axon terminals targeted CeA neurons that expressed CRF. Triple label immunofluorescence microscopy revealed that DOPR and CRF were co-localized in common cellular profiles that were in close proximity to NE-containing fibers in both subregions. These anatomical results indicate significant interactions between DOPR and CRF in this critical limbic region and reveal that NE is poised to regulate these peptidergic systems in the amygdala. Functional studies were performed to determine if activation of DOPR could inhibit the anxiety produced by elevation of NE in the amygdala using the pharmacological stressor yohimbine. Administration of the DOPR agonist, SNC80, significantly attenuated elevated anxiogenic behaviors produced by yohimbine as measured in the rat on the elevated zero maze. Taken together, results from this study demonstrate the convergence of three important systems, NE, CRF, and DOPR, in the amygdala and provide insight into their functional role in modulating stress and anxiety responses.


Asunto(s)
Ansiedad/fisiopatología , Complejo Nuclear Basolateral/metabolismo , Complejo Nuclear Basolateral/ultraestructura , Núcleo Amigdalino Central/metabolismo , Núcleo Amigdalino Central/ultraestructura , Hormona Liberadora de Corticotropina/metabolismo , Receptores Opioides delta/metabolismo , Neuronas Adrenérgicas/citología , Neuronas Adrenérgicas/metabolismo , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/ultraestructura , Animales , Benzamidas/administración & dosificación , Masculino , Neuronas/metabolismo , Neuronas/ultraestructura , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Piperazinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/agonistas
16.
eNeuro ; 2(6)2015.
Artículo en Inglés | MEDLINE | ID: mdl-26634226

RESUMEN

Stress increases vulnerability to psychiatric disorders, partly by affecting brain monoamine systems, such as the locus coeruleus (LC)-norepinephrine system. During stress, LC activity is coregulated by corticotropin-releasing factor (CRF) and endogenous opioids. This study identified neural circuitry that regulates LC activity of intruder rats during the resident-intruder model of social stress. LC afferents were retrogradely labeled with Fluorogold (FG) and rats were subjected to one or five daily exposures to an aggressive resident. Sections through the nucleus paragigantocellularis (PGi) and central amygdalar nucleus (CNA), major sources of enkephalin (ENK) and CRF LC afferents, respectively, were immunocytochemically processed to detect c-fos, FG, and CRF or ENK. In response to a single exposure, intruder rats assumed defeat with a relatively short latency (SL). LC neurons, PGI-ENK LC afferents, and CNA-CRF LC afferents were activated in these rats as indicated by increased c-fos expression. With repeated stress, rats exhibited either a SL or long latency (LL) to defeat and these strategies were associated with distinct patterns of neuronal activation. In SL rats, LC neurons were activated, as were CNA-CRF LC afferents but not PGI-ENK LC afferents. LL rats had an opposite pattern, maintaining activation of PGi-ENK LC afferents but not CNA-CRF LC afferents or LC neurons. Together, these results indicate that the establishment of different coping strategies to social stress is associated with changes in the circuitry that regulates activity of the brain norepinephrine system. This may underlie differential vulnerability to the consequences of social stress that characterize these different coping strategies.


Asunto(s)
Adaptación Psicológica/fisiología , Amígdala del Cerebelo/metabolismo , Hormona Liberadora de Corticotropina/análisis , Locus Coeruleus/citología , Estrés Psicológico/metabolismo , Animales , Conducta Animal/fisiología , Encefalinas/análisis , Masculino , Neuronas/metabolismo , Norepinefrina/análisis , Ratas Long-Evans
17.
Eur J Neurosci ; 40(8): 3202-14, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25131562

RESUMEN

Endocannabinoids acting at the cannabinoid type 1 receptor (CB1R) are known to regulate attention, cognition and mood. Previous studies have shown that, in the rat medial prefrontal cortex (mPFC), CB1R agonists increase norepinephrine release, an effect that may be attributed, in part, to CB1Rs localised to noradrenergic axon terminals. The present study was aimed at further characterising functional interactions between CB1R and adrenergic receptor (AR) systems in the mPFC using in vitro intracellular electrophysiology and high-resolution neuroanatomical techniques. Whole-cell patch-clamp recordings of layer V/VI cortical pyramidal neurons in rats revealed that both acute and chronic treatment with the synthetic CB1R agonist WIN 55,212-2 blocked elevations in cortical pyramidal cell excitability and increases in input resistance evoked by the α2-adrenergic receptor (α2-AR) agonist clonidine, suggesting a desensitisation of α2-ARs. These CB1R-α2-AR interactions were further shown to be both action potential- and gamma-aminobutyric acid-independent. To better define sites of cannabinoid-AR interactions, we localised α2A-adrenergic receptors (α2A-ARs) in a genetically modified mouse that expressed a hemoagglutinin (HA) tag downstream of the α2A-AR promoter. Light and electron microscopy indicated that HA-α2A-AR was distributed in axon terminals and somatodendritic processes especially in layer V of the mPFC. Triple-labeling immunocytochemistry revealed that α2A-AR and CB1R were localised to processes that contained dopamine-ß-hydroxylase, a marker of norepinephrine. Furthermore, HA-α2A-AR was localised to processes that were directly apposed to CB1R. These findings suggest multiple sites of interaction between cortical cannabinoid-adrenergic systems that may contribute to understanding the effect of cannabinoids on executive functions and mood.


Asunto(s)
Corteza Prefrontal/fisiología , Células Piramidales/fisiología , Receptor Cannabinoide CB1/fisiología , Receptores Adrenérgicos alfa 2/fisiología , Potenciales de Acción/efectos de los fármacos , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Benzoxazinas/farmacología , Clonidina/farmacología , Técnicas de Sustitución del Gen , Masculino , Ratones , Morfolinas/farmacología , Naftalenos/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/ultraestructura , Células Piramidales/efectos de los fármacos , Células Piramidales/ultraestructura , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/agonistas , Receptores Adrenérgicos alfa 2/análisis , Receptores Adrenérgicos alfa 2/genética
18.
Autophagy ; 10(7): 1193-211, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24813396

RESUMEN

Although autophagic pathways are essential to developmental processes, many questions still remain regarding the initiation signals that regulate autophagy in the context of differentiation. To address these questions we studied the ocular lens, as the programmed elimination of nuclei and organelles occurs in a precisely regulated spatiotemporal manner to form the organelle-free zone (OFZ), a characteristic essential for vision acuity. Here, we report our discovery that inactivation of MAPK/JNK induces autophagy for formation of the OFZ through its regulation of MTORC1, where MAPK/JNK signaling is required for both MTOR activation and RPTOR/RAPTOR phosphorylation. Autophagy pathway proteins including ULK1, BECN1/Beclin 1, and MAP1LC3B2/LC3B-II were upregulated in the presence of inhibitors to either MAPK/JNK or MTOR, inducing autophagic loss of organelles to form the OFZ. These results reveal that MAPK/JNK is a positive regulator of MTORC1 signaling and its developmentally regulated inactivation provides an inducing signal for the coordinated autophagic removal of nuclei and organelles required for lens function.


Asunto(s)
Autofagia , Diferenciación Celular , Núcleo Celular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Cristalino/patología , Sistema de Señalización de MAP Quinasas , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Proteínas Aviares/metabolismo , Núcleo Celular/ultraestructura , Embrión de Pollo , Vesículas Citoplasmáticas/metabolismo , Vesículas Citoplasmáticas/ultraestructura , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Cristalino/embriología , Cristalino/ultraestructura , Diana Mecanicista del Complejo 1 de la Rapamicina , Modelos Biológicos , Factores de Tiempo
19.
Artículo en Inglés | MEDLINE | ID: mdl-24333843

RESUMEN

Opiate addiction is a devastating health problem, with approximately 2million people currently addicted to heroin or non-medical prescription opiates in the United States alone. In neurons, adaptations in cell signaling cascades develop following opioid actions at the mu opioid receptor (MOR). A novel putative target for intervention involves interacting proteins that may regulate trafficking of MOR. Morphine has been shown to induce a re-distribution of a MOR-interacting protein Wntless (WLS, a transport molecule necessary for secretion of neurotrophic Wnt proteins), from cytoplasmic to membrane compartments in rat striatal neurons. Given its opiate-sensitivity and its well-characterized molecular and cellular adaptations to morphine exposure, we investigated the anatomical distribution of WLS and MOR in the rat locus coeruleus (LC)-norepinephrine (NE) system. Dual immunofluorescence microscopy was used to test the hypothesis that WLS is localized to noradrenergic neurons of the LC and that WLS and MOR co-exist in common LC somatodendritic processes, providing an anatomical substrate for their putative interactions. We also hypothesized that morphine would influence WLS distribution in the LC. Rats received saline, morphine or the opiate agonist [d-Ala2, N-Me-Phe4, Gly-ol5]-enkephalin (DAMGO), and tissue sections through the LC were processed for immunogold-silver detection of WLS and MOR. Statistical analysis showed a significant re-distribution of WLS to the plasma membrane following morphine treatment in addition to an increase in the proximity of gold-silver labels for MOR and WLS. Following DAMGO treatment, MOR and WLS were predominantly localized within the cytoplasmic compartment when compared to morphine and control. In a separate cohort of rats, brains were obtained from saline-treated or heroin self-administering male rats for pulldown co-immunoprecipitation studies. Results showed an increased association of WLS and MOR following heroin exposure. As the LC-NE system is important for cognition as well as decisions underlying substance abuse, adaptations in WLS trafficking and expression may play a role in modulating MOR function in the LC and contribute to the negative sequelae of opiate exposure on executive function.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Locus Coeruleus/citología , Morfina/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides mu/metabolismo , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/ultraestructura , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/antagonistas & inhibidores , Analgésicos Opioides/farmacología , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Heroína/administración & dosificación , Heroína/farmacología , Infusiones Intraventriculares , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/ultraestructura , Masculino , Morfina/administración & dosificación , Morfina/antagonistas & inhibidores , Unión Proteica , Transporte de Proteínas/efectos de los fármacos , Ratas , Autoadministración
20.
Antioxidants (Basel) ; 3(2): 414-38, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-26784879

RESUMEN

HIV-1 trans-acting protein Tat, an essential protein for viral replication, is a key mediator of neurotoxicity. If Tat oxidant injury and neurotoxicity have been described, consequent neuroinflammation is less understood. Rat caudate-putamens (CPs) were challenged with Tat, with or without prior rSV40-delivered superoxide dismutase or glutathione peroxidase. Tat injection caused oxidative stress. Administration of Tat in the CP induced an increase in numbers of Iba-1- and CD68-positive cells, as well as an infiltration of astrocytes. We also tested the effect of more protracted Tat exposure on neuroinflammation using an experimental model of chronic Tat exposure. SV(Tat): a recombinant SV40-derived gene transfer vector was inoculated into the rat CP, leading to chronic expression of Tat, oxidative stress, and ongoing apoptosis, mainly located in neurons. Intra-CP SV(Tat) injection induced an increase in microglia and astrocytes, suggesting that protracted Tat production increased neuroinflammation. SV(SOD1) or SV(GPx1) significantly reduced neuroinflammation following Tat administration into the CP. Thus, Tat-induced oxidative stress, CNS injury, neuron loss and inflammation may be mitigated by antioxidant gene delivery.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...