Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39060161

RESUMEN

Cannabis and its products have been used for centuries for both medicinal and recreational purposes. The recent widespread legalization of cannabis has vastly expanded its use in the United States across all demographics except for adolescents. Meanwhile decades of research have advanced our knowledge of cannabis pharmacology and particularly of the endocannabinoid system with which the components of cannabis interact. This research has revealed multiple targets and approaches for manipulating the system for therapeutic use and to ameliorate cannabis toxicity or cannabis use disorder. Research has also led to new questions that underscore the potential risks of its widespread use, particularly the enduring consequences of exposure during critical windows of brain development or for consumption of large daily doses of cannabis with high content D9 tetrahydrocannabinol (THC). Here we highlight current neuroscience research on cannabis that has shed light on therapeutic opportunities and potential adverse consequences of misuse and point to gaps in knowledge that can guide future research. Significance Statement Cannabis use has escalated with its increased availability. Here we highlight the challenges of cannabis research and the gaps in our knowledge of cannabis pharmacology and of the endocannabinoid system that it targets. Future research that addresses these gaps is needed so that the endocannabinoid system can be leveraged for safe and effective use.

2.
Artículo en Inglés | MEDLINE | ID: mdl-37947883

RESUMEN

The prevention and treatment of addiction (moderate to severe substance use disorder-SUD) have remained challenging because of the dynamic and complex interactions between multiple biological and social determinants that shape SUD. The pharmacological landscape is ever changing and the use of multiple drugs is increasingly common, requiring an unraveling of pharmacological interactions to understand the effects. There are different stages in the trajectory from drug use to addiction that are characterized by distinct cognitive and emotional features. These are directed by different neurobiological processes that require identification and characterization including those that underlie the high co-morbidity with other disorders. Finally, there is substantial individual variability in the susceptibility to develop SUD because there are multiple determinants, including genetics, sex, developmental trajectories and times of drug exposures, and psychosocial and environmental factors including commercial determinants that influence drug availability. Elucidating how these factors interact to determine risk is essential for identifying the biobehavioral basis of addiction and developing prevention and treatment strategies. Basic research is tasked with addressing each of these challenges. The recent proliferation of technological advances that allow for genetic manipulation, visualization of molecular reactions and cellular activity in vivo, multiscale whole brain mapping across the life span, and the mining of massive data sets including multimodality human brain imaging are accelerating our ability to understand how the brain functions and how drugs influence it. Here, we highlight how the application of these tools to the study of addiction promises to illuminate its neurobiological basis and guide strategies for prevention and treatment.

3.
J Neurosci ; 41(21): 4550-4555, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-33926994

RESUMEN

Cholinergic regulation of hippocampal circuit activity has been an active area of neurophysiological research for decades. The prominent cholinergic innervation of intrinsic hippocampal circuitry, potent effects of cholinomimetic drugs, and behavioral responses to cholinergic modulation of hippocampal circuitry have driven investigators to discover diverse cellular actions of acetylcholine in distinct sites within hippocampal circuitry. Further research has illuminated how these actions organize circuit activity to optimize encoding of new information, promote consolidation, and coordinate this with recall of prior memories. The development of the hippocampal slice preparation was a major advance that accelerated knowledge of how hippocampal circuits functioned and how acetylcholine modulated these circuits. Using this preparation in the early 1980s, we made a serendipitous finding of a novel presynaptic inhibitory effect of acetylcholine on Schaffer collaterals, the projections from CA3 pyramidal neurons to dendrites of CA1 pyramidal cells. We characterized this effect at cellular and pharmacological levels, published the findings in the first volume of the Journal of Neuroscience, and proceeded to pursue other scientific directions. We were surprised and thrilled to see that, nearly 40 years later, the paper is still being cited and downloaded because the data became an integral piece of the foundation of the science of cholinergic regulation of hippocampal function in learning and memory. This Progressions article is a story of how single laboratory findings evolve through time to be confirmed, challenged, and reinterpreted by other laboratories to eventually become part of the basis of fundamental concepts related to important brain functions.


Asunto(s)
Acetilcolina/metabolismo , Hipocampo/fisiología , Neurología/historia , Técnicas de Cultivo de Órganos/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Técnicas de Cultivo de Órganos/métodos
4.
J Neurosci ; 41(34): 7314-7325, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34193553

RESUMEN

Lower urinary tract or voiding disorders are prevalent across all ages and affect >40% of adults over 40 years old, leading to decreased quality of life and high health care costs. The pontine micturition center (PMC; i.e., Barrington's nucleus) contains a large population of neurons that localize the stress-related neuropeptide, corticotropin-releasing hormone (CRH) and project to neurons in the spinal cord to regulate micturition. How the PMC and CRH-expressing neurons in the PMC control volitional micturition is of critical importance for human voiding disorders. To investigate the specific role of CRH in the PMC, neurons in the PMC-expressing CRH were optogenetically activated during in vivo cystometry in unanesthetized mice of either sex. Optogenetic activation of CRH-PMC neurons led to increased intermicturition interval and voided volume, similar to the altered voiding phenotype produced by social stress. Female mice showed a significantly more pronounced phenotype change compared with male mice. These effects were eliminated by CRH-receptor 1 antagonist pretreatment. Optogenetic inhibition of CRH-PMC neurons led to an altered voiding phenotype characterized by more frequent voids and smaller voided volumes. Last, in a cyclophosphamide cystitis model of bladder overactivity, optogenetic activation of CRH-PMC neurons returned the voiding pattern to normal. Collectively, our findings demonstrate that CRH from PMC spinal-projecting neurons has an inhibitory function on micturition and is a potential therapeutic target for human disease states, such as voiding postponement, urinary retention, and underactive or overactive bladder.SIGNIFICANCE STATEMENT The pontine micturition center (PMC), which is a major regulator of volitional micturition, is neurochemically heterogeneous, and excitatory neurotransmission derived from PMC neurons is thought to mediate the micturition reflex. In the present study, using optogenetic manipulation of CRH-containing neurons in double-transgenic mice, we demonstrate that CRH, which is prominent in PMC-spinal projections, has an inhibitory function on volitional micturition. Moreover, engaging this inhibitory function of CRH can ameliorate bladder hyperexcitability induced by cyclophosphamide in a model of cystitis. The data underscore CRH as a novel target for the treatment of voiding dysfunctions, which are highly prevalent disease processes in children and adults.


Asunto(s)
Núcleo de Barrington/fisiología , Hormona Liberadora de Corticotropina/metabolismo , Micción/fisiología , Vías Aferentes/fisiología , Animales , Proteínas Arqueales/genética , Núcleo de Barrington/citología , Channelrhodopsins/genética , Hormona Liberadora de Corticotropina/genética , Ciclofosfamida/toxicidad , Cistitis/inducido químicamente , Cistitis/tratamiento farmacológico , Cistitis/fisiopatología , Femenino , Genes Reporteros/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/fisiología , Optogenética , Fotoquímica , Proteínas Recombinantes/genética , Médula Espinal/fisiología , Urodinámica , Volición
5.
Mol Psychiatry ; 26(1): 41-50, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33188253

RESUMEN

Interventions to address the U.S. opioid crisis primarily target opioid use, misuse, and addiction, but because the opioid crisis includes multiple substances, the opioid specificity of interventions may limit their ability to address the broader problem of polysubstance use. Overlap of opioids with other substances ranges from shifts among the substances used across the lifespan to simultaneous co-use of substances that span similar and disparate pharmacological categories. Evidence suggests that nonmedical opioid users quite commonly use other drugs, and this polysubstance use contributes to increasing morbidity and mortality. Reasons for adding other substances to opioids include enhancement of the high (additive or synergistic reward), compensation for undesired effects of one drug by taking another, compensation for negative internal states, or a common predisposition that is related to all substance consumption. But consumption of multiple substances may itself have unique effects. To achieve the maximum benefit, addressing the overlap of opioids with multiple other substances is needed across the spectrum of prevention and treatment interventions, overdose reversal, public health surveillance, and research. By addressing the multiple patterns of consumption and the reasons that people mix opioids with other substances, interventions and research may be enhanced.


Asunto(s)
Epidemia de Opioides , Trastornos Relacionados con Opioides/epidemiología , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Conducta Adictiva/epidemiología , Interacciones Farmacológicas , Sobredosis de Droga/epidemiología , Sobredosis de Droga/prevención & control , Sobredosis de Droga/terapia , Humanos , Trastornos Relacionados con Opioides/tratamiento farmacológico , Trastornos Relacionados con Opioides/prevención & control , Estados Unidos/epidemiología
6.
Mol Pharmacol ; 98(4): 389-391, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32660966

RESUMEN

The International Narcotics Research Conference (INRC) has a rich history of uniting the most creative minds across the fields of chemistry, pharmacology, physiology, and behavior in the study of opioids. The Conference has been a forum for sharing knowledge, discussing controversies, introducing innovative research, and announcing landmark discoveries. In this perspective for the Special Issue commemorating the 50th anniversary of the Conference we briefly highlight how the INRC has guided the evolution of opioid research and how new tools, models, and approaches are facilitating our ability to achieve the goals of preventing and treating opioid use disorder. SIGNIFICANCE STATEMENT: This perspective highlights the important role that the International Narcotics Research Conference has played in the evolution of opioid research and emphasizes how technological advances are facilitating research toward the goals of preventing and treating opioid use disorder.


Asunto(s)
Analgésicos Opioides/efectos adversos , Trastornos Relacionados con Opioides/prevención & control , Dolor/tratamiento farmacológico , Analgésicos Opioides/uso terapéutico , Investigación Biomédica , Encéfalo/metabolismo , Encéfalo/fisiopatología , Congresos como Asunto , Humanos , Trastornos Relacionados con Opioides/tratamiento farmacológico , Trastornos Relacionados con Opioides/metabolismo , Dolor/fisiopatología , Receptores Opioides/metabolismo
7.
J Neurosci ; 37(45): 10848-10854, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118214

RESUMEN

The brain processes information from the periphery and regulates visceral and immune activity to maintain internal homeostasis, optimally respond to a dynamic external environment, and integrate these functions with ongoing behavior. In addition to its relevance for survival, this integration underlies pathology as evidenced by diseases exhibiting comorbid visceral and psychiatric symptoms. Advances in neuroanatomical mapping, genetically specific neuronal manipulation, and neural network recording are overcoming the challenges of dissecting complex circuits that underlie this integration and deciphering their function. Here we focus on reciprocal communication between the brain and urological, gastrointestinal, and immune systems. These studies are revealing how autonomic activity becomes integrated into behavior as part of a social strategy, how the brain regulates innate immunity in response to stress, and how drugs impact emotion and gastrointestinal function. These examples highlight the power of the functional organization of circuits at the interface of the brain and periphery.


Asunto(s)
Sistema Nervioso Central/fisiología , Homeostasis/fisiología , Inmunidad Humoral/fisiología , Red Nerviosa/fisiología , Animales , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/fisiología , Humanos , Red Nerviosa/inmunología
8.
Cereb Cortex ; 27(1): 244-253, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28013234

RESUMEN

Stress is implicated in psychiatric illnesses that are characterized by impairments in cognitive functions that are mediated by the medial prefrontal cortex (mPFC). Because sex and age determine stress vulnerability, the effects of repeated social stress occurring during early adolescence, mid-adolescence, or adulthood on the cellular properties of male and female rat mPFC Layer V neurons in vitro were examined. Repeated resident-intruder stress produced age- and sex-specific effects on mPFC intrinsic and synaptic excitability. Mid-adolescents were particularly vulnerable to effects on intrinsic excitability. The maximum number of action potentials (APs) evoked by increasing current intensity was robustly decreased in stressed male and female mid-adolescent rats compared with age-matched controls. These effects were associated with stress-induced changes in AP half-width, amplitude, threshold, and input resistance. Social stress at all ages generally decreased synaptic excitability by decreasing the amplitude of spontaneous excitatory postsynaptic potentials. The results suggest that whereas social stress throughout life can diminish the influence of afferents driving the mPFC, social stress during mid-adolescence additionally affects intrinsic characteristics of mPFC neurons that determine excitability. The depressant effects of social stress on intrinsic and synaptic mPFC neurons may underlie its ability to affect executive functions and emotional responses, particularly during adolescence.


Asunto(s)
Envejecimiento , Excitabilidad Cortical , Corteza Prefrontal/fisiopatología , Memoria Implícita , Medio Social , Estrés Psicológico/fisiopatología , Transmisión Sináptica/fisiología , Animales , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Estigma Social
9.
Addict Biol ; 21(1): 111-24, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25262980

RESUMEN

Both the opioid antagonist naltrexone and corticotropin-releasing factor type-1 receptor (CRF-R1) antagonists have been investigated for the treatment of alcoholism. The current study examines the combination of naltrexone and CP154526 to reduce intermittent access ethanol drinking [intermittent access to alcohol (IAA)] in C57BL/6J male mice, and if these compounds reduce drinking via serotonergic mechanisms in the dorsal raphe nucleus (DRN). Systemic injections and chronic intracerebroventricular infusions of naltrexone, CP154526 or CP376395 transiently decreased IAA drinking. Immunohistochemistry revealed CRF-R1 or µ-opioid receptor immunoreactivity was co-localized in tryptophan hydroxylase (TPH)-immunoreactive neurons as well as non-TPH neurons in the DRN. Mice with a history of IAA or continuous access to alcohol were microinjected with artificial cerebral spinal fluid, naltrexone, CP154526 or the combination into the DRN or the median raphe nucleus (MRN). Either intra-DRN naltrexone or CP154526 reduced IAA in the initial 2 hours of fluid access, but the combination did not additively suppress IAA, suggesting a common mechanism via which these two compounds affect intermittent drinking. These alcohol-reducing effects were localized to the DRN of IAA drinkers, as intra-MRN injections only significantly suppressed water drinking, and continuous access drinkers were not affected by CRF-R1 antagonism. Extracellular serotonin was measured in the medial prefrontal cortex (mPFC) using in vivo microdialysis after intra-DRN microinjections in another group of mice. Intra-DRN CP154526 increased serotonin impulse flow to the mPFC while naltrexone did not. This suggests the mPFC may not be an essential location to intermittent drinking, as evidenced by different effects on serotonin signaling to the forebrain yet similar behavioral findings.


Asunto(s)
Consumo de Bebidas Alcohólicas , Conducta Animal/efectos de los fármacos , Depresores del Sistema Nervioso Central/administración & dosificación , Núcleo Dorsal del Rafe , Etanol/administración & dosificación , Antagonistas de Narcóticos/farmacología , Corteza Prefrontal/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores Opioides mu/antagonistas & inhibidores , Aminopiridinas/farmacología , Animales , Inmunohistoquímica , Infusiones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Microdiálisis , Naltrexona/farmacología , Corteza Prefrontal/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores Opioides mu/metabolismo , Autoadministración , Serotonina/metabolismo , Triptófano Hidroxilasa/metabolismo
10.
Front Neuroendocrinol ; 35(3): 303-19, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24726661

RESUMEN

Stress is associated with the onset and severity of several psychiatric disorders that occur more frequently in women than men, including posttraumatic stress disorder (PTSD) and depression. Patients with these disorders present with dysregulation of several stress response systems, including the neuroendocrine response to stress, corticolimbic responses to negatively valenced stimuli, and hyperarousal. Thus, sex differences within their underlying circuitry may explain sex biases in disease prevalence. This review describes clinical studies that identify sex differences within the activity of these circuits, as well as preclinical studies that demonstrate cellular and molecular sex differences in stress responses systems. These studies reveal sex differences from the molecular to the systems level that increase endocrine, emotional, and arousal responses to stress in females. Exploring these sex differences is critical because this research can reveal the neurobiological underpinnings of vulnerability to stress-related psychiatric disorders and guide the development of novel pharmacotherapies.


Asunto(s)
Trastornos Mentales , Estrés Psicológico , Animales , Nivel de Alerta/fisiología , Encéfalo , Hormona Liberadora de Corticotropina/metabolismo , Humanos , Caracteres Sexuales
11.
Mol Pharmacol ; 83(4): 737-45, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23239826

RESUMEN

Sex differences in the prevalence or severity of many diseases and in the response to pharmacological agents are well recognized. Elucidating the biologic bases of these differences can advance our understanding of the pathophysiology of disease and facilitate the development of treatments. Despite the importance to medicine, this has been an area of limited research. Here, we review physiologic, cellular, and molecular findings supporting the idea that there are sex differences in receptor signaling and trafficking that can be determinants of pathology. The focus is on the receptor for corticotropin-releasing factor (CRF), the orchestrator of the stress response, which has been implicated in diverse stress-related diseases that show a female prevalence. Data are reviewed that show sex differences in the association of the CRF receptor (CRF1) with the Gs protein and ß-arrestin 2 that would render females more responsive to acute stress and less able to adapt to chronic stress as a result of compromised CRF1 internalization. Because ß-arrestin 2 serves to link CRF1 to Gs-independent signaling pathways, this sex-biased signaling is proposed to result in distinct cellular responses to stress that are translated to different physiologic and behavioral coping mechanisms and that can have different pathologic consequences. Because stress has been implicated in diverse medical and psychiatric diseases, these sex differences in CRF1 signaling could explain sex differences in a multitude of disorders. The possibility that analogous sex differences may occur with other G-protein-coupled receptors underscores the impact of this effect and is discussed.


Asunto(s)
Modelos Moleculares , Receptores de Hormona Liberadora de Corticotropina/fisiología , Caracteres Sexuales , Transducción de Señal/fisiología , Estrés Psicológico/metabolismo , Animales , Hormona Liberadora de Corticotropina/química , Hormona Liberadora de Corticotropina/metabolismo , Hormona Liberadora de Corticotropina/fisiología , Femenino , Humanos , Masculino , Trastornos Mentales/metabolismo , Trastornos Mentales/psicología , Modelos Químicos , Transporte de Proteínas/fisiología , Receptores de Hormona Liberadora de Corticotropina/química , Estrés Psicológico/psicología
12.
Eur J Neurosci ; 37(6): 901-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23279008

RESUMEN

Data from preclinical and clinical studies have implicated the norepinephrine system in the development and maintenance of post-traumatic stress disorder. The primary source of norepinephrine in the forebrain is the locus coeruleus (LC); however, LC activity cannot be directly measured in humans, and previous research has often relied upon peripheral measures of norepinephrine to infer changes in central LC-norepinephrine function. To directly assess LC-norepinephrine function, we measured single-unit activity of LC neurons in a validated rat model of post-traumatic stress disorder - single prolonged stress (SPS). We also examined tyrosine hydroxylase mRNA levels in the LC of SPS and control rats as an index of norepinephrine utilisation. For electrophysiological recordings, 92 LC neurons were identified from 19 rats (SPS, 12; control, 7), and spontaneous and evoked responses to a noxious event (paw compression) were recorded. Baseline and restraint stress-evoked tyrosine hydroxylase mRNA expression levels were measured in SPS and control rats (n = 16 per group) in a separate experiment. SPS rats showed lower spontaneous activity but higher evoked responses, leading to an enhanced signal-to-noise ratio of LC neurons, accompanied by impaired recovery from post-stimulus inhibition. In concert, tyrosine hydroxylase mRNA expression in the LC of SPS rats tended to be lower at baseline, but was exaggerated following restraint stress. These data demonstrate persistent changes in LC function following stress/trauma in a rat model of post-traumatic stress, as measured by differences in both the electrophysiological properties of LC neurons and tyrosine hydroxylase mRNA transcription.


Asunto(s)
Locus Coeruleus/metabolismo , Norepinefrina/metabolismo , Estrés Psicológico/fisiopatología , Potenciales de Acción , Animales , Locus Coeruleus/citología , Locus Coeruleus/fisiopatología , Masculino , Inhibición Neural , Neuronas/metabolismo , Neuronas/fisiología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Restricción Física , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/fisiopatología , Estrés Psicológico/metabolismo , Transcripción Genética , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
13.
Am J Physiol Regul Integr Comp Physiol ; 304(11): R940-50, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23552576

RESUMEN

Barrington's nucleus, in the pons, regulates micturition through spinal projections to preganglionic parasympathetic neurons. The stress neuropeptide CRF is prominent in these projections and has an inhibitory influence. Social stress in rats causes urinary retention and abnormal urodynamics resembling those produced by partial bladder outlet obstruction (pBOO), and this is associated with CRF upregulation in Barrington's nucleus. Here, we examined the role of CRF in social stress- and pBOO-induced urodynamic dysfunction by assessing the ability of a CRF1 receptor antagonist to alter these effects. Male rats exposed to repeated resident-intruder stress were administered vehicle or a CRF1 antagonist (NBI-30775) daily prior to the stress. Urodynamic function was recorded in the unanesthetized state 72 h after the final stress. NBI-30775 prevented the increased intermicturition interval, micturition volume, and bladder capacity produced by social stress, but not the increase in CRF expression in Barrington's nucleus neurons. The urinary dysfunction was also partly prevented by shRNA targeting of CRF in Barrington's nucleus, suggesting that stress-induced urinary dysfunction results, in part, from CRF upregulation in Barrington's nucleus and enhanced postsynaptic effects in the spinal cord. Finally, NBI-30775 improved urodynamic function of rats that had pBOO of 2-wk duration when administered daily during the second week but did not block the increase in CRF expression in Barrington's nucleus neurons. These findings implicate a role for Barrington's nucleus CRF in stress- and pBOO-induced urodynamic changes and suggest that CRF1 antagonists may be useful therapeutic agents for the treatment of urinary dysfunction.


Asunto(s)
Pirimidinas/uso terapéutico , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Estrés Psicológico/orina , Obstrucción del Cuello de la Vejiga Urinaria/tratamiento farmacológico , Urodinámica/efectos de los fármacos , Animales , Hormona Liberadora de Corticotropina/biosíntesis , Dependovirus/genética , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Masculino , Puente/fisiología , ARN/análisis , ARN/biosíntesis , ARN Interferente Pequeño/genética , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Medio Social , Estrés Psicológico/psicología , Obstrucción del Cuello de la Vejiga Urinaria/fisiopatología
14.
J Neurosci ; 31(40): 14436-49, 2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-21976528

RESUMEN

Because overactivation of the hypothalamic-pituitary-adrenal (HPA) axis occurs in Alzheimer's disease (AD), dysregulation of stress neuromediators may play a mechanistic role in the pathophysiology of AD. However, the effects of stress on tau phosphorylation are poorly understood, and the relationship between corticosterone and corticotropin-releasing factor (CRF) on both ß-amyloid (Aß) and tau pathology remain unclear. Therefore, we first established a model of chronic stress, which exacerbates Aß accumulation in Tg2576 mice and then extended this stress paradigm to a tau transgenic mouse model with the P301S mutation (PS19) that displays tau hyperphosphorylation, insoluble tau inclusions and neurodegeneration. We show for the first time that both Tg2576 and PS19 mice demonstrate a heightened HPA stress profile in the unstressed state. In Tg2576 mice, 1 month of restraint/isolation (RI) stress increased Aß levels, suppressed microglial activation, and worsened spatial and fear memory compared with nonstressed mice. In PS19 mice, RI stress promoted tau hyperphosphorylation, insoluble tau aggregation, neurodegeneration, and fear-memory impairments. These effects were not mimicked by chronic corticosterone administration but were prevented by pre-stress administration of a CRF receptor type 1 (CRF(1)) antagonist. The role for a CRF(1)-dependent mechanism was further supported by the finding that mice overexpressing CRF had increased hyperphosphorylated tau compared with wild-type littermates. Together, these results implicate HPA dysregulation in AD neuropathogenesis and suggest that prolonged stress may increase Aß and tau hyperphosphorylation. These studies also implicate CRF in AD pathophysiology and suggest that pharmacological manipulation of this neuropeptide may be a potential therapeutic strategy for AD.


Asunto(s)
Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Degeneración Nerviosa/patología , Receptores de Hormona Liberadora de Corticotropina/biosíntesis , Estrés Psicológico/patología , Tauopatías/patología , Animales , Enfermedad Crónica , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/metabolismo , Femenino , Sistema Hipotálamo-Hipofisario/patología , Sistema Hipotálamo-Hipofisario/fisiología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Degeneración Nerviosa/etiología , Degeneración Nerviosa/metabolismo , Sistema Hipófiso-Suprarrenal/patología , Sistema Hipófiso-Suprarrenal/fisiología , Estrés Psicológico/complicaciones , Estrés Psicológico/metabolismo , Tauopatías/etiología , Tauopatías/metabolismo , Proteínas tau/biosíntesis
15.
Eur J Neurosci ; 36(10): 3356-64, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22882375

RESUMEN

The stress-related neuropeptide, corticotropin-releasing factor (CRF), is prominent in neurons of the pontine micturition center, Barrington's nucleus. These neurons co-innervate spinal preganglionic neurons that control the bladder, and locus coeruleus (LC) neurons that provide norepinephrine innervation throughout the brain. Adeno-associated viral (AAV) vector-mediated transfer of CRF cDNA was used to increase CRF expression in Barrington's nucleus neurons and investigate the impact of a gain of function in Barrington's nucleus spinal and LC projections. AAV transfer of the reverse CRF cDNA sequence served as the control. Bladder urodynamics and behavior were assessed 4 weeks after vector injection into Barrington's nucleus. Rats with bilateral injections of AAV-CRF cDNA into Barrington's nucleus had immunohistochemical evidence of CRF overexpression in neurons and transport to the spinal cord and LC. The bladder : body weight ratio was greater and micturition pressure was less in these rats compared with controls, consistent with an inhibitory influence on bladder function. Other indices of urodynamic function were not altered. CRF innervation of the LC was increased in rats with bilateral Barrington's nucleus injections of AAV-CRF cDNA, and this was associated with increased burying behavior, an endpoint of LC activation by CRF. The results provide immunohistochemical evidence for viral vector-induced CRF overexpression in Barrington's nucleus neurons and underscore the ability of AAV vector-mediated transfer to increase CRF function in selective circuits. The findings support an inhibitory influence of CRF in Barrington's nucleus regulation of the bladder and an excitatory influence on the brain norepinephrine system that translates to behavioral activation.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Locus Coeruleus/fisiología , Puente/fisiología , Vejiga Urinaria/fisiología , Micción/fisiología , Urodinámica/fisiología , Animales , Conducta Animal , Peso Corporal , Hormona Liberadora de Corticotropina/genética , Dependovirus/genética , Vectores Genéticos , Masculino , Ratones , Neuronas/metabolismo , Puente/citología , Puente/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Médula Espinal/fisiología , Transducción Genética , Regulación hacia Arriba , Vejiga Urinaria/inervación
16.
Cell Mol Neurobiol ; 32(5): 709-23, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22488525

RESUMEN

Women are twice as likely as men to suffer from stress-related psychiatric disorders, like unipolar depression and post-traumatic stress disorder. Although the underlying neural mechanisms are not well characterized, the pivotal role of stress in the onset and severity of these diseases has led to the idea that sex differences in stress responses account for this sex bias. Corticotropin-releasing factor (CRF) orchestrates stress responses by acting both as a neurohormone to initiate the hypothalamic-pituitary-adrenal (HPA) axis and as a neuromodulator in the brain. One target of CRF modulation is the locus coeruleus (LC)-norepinephrine system, which coordinates arousal components of the stress response. Hypersecretion of CRF and dysregulation of targets downstream from CRF, such as the HPA axis and LC-norepinephrine system, are characteristic features of many stress-related psychiatric diseases, suggesting a causal role for CRF and its targets in the development of these disorders. This review will describe sex differences in CRF and the LC-norepinephrine system that can increase stress sensitivity in females, making them vulnerable to stress-related disorders. Evidence for gonadal hormone regulation of hypothalamic CRF is discussed as an effect that can lead to increased HPA axis activity in females. Sex differences in the structure of LC neurons that create the potential for hyperarousal in response to emotional stimuli are described. Finally, sex differences at the molecular level of the CRF(1) receptor that make the LC-norepinephrine system more reactive in females are reviewed. The implications of these sex differences for the treatment of stress-related psychiatric disorders also will be discussed.


Asunto(s)
Caracteres Sexuales , Transducción de Señal , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Humanos , Locus Coeruleus/metabolismo , Masculino , Receptores de Hormona Liberadora de Corticotropina/metabolismo
17.
J Neurosci ; 30(48): 16149-59, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21123561

RESUMEN

The transcription factor cAMP response element-binding protein (CREB) is required for stress- but not drug-induced reinstatement of cocaine conditioned place preference. To reveal the neural circuitry associated with this CREB dependence, we injected a retrograde tracer into the ventral tegmental area (VTA) and identified afferents that were activated after stress or cocaine exposure in both naive and cocaine-conditioned mice. Neuronal activation, as assessed by Fos expression, was greatly reduced in the dorsal and ventral bed nucleus of the stria terminalis (BNST), lateral septum, and nucleus accumbens shell in mice lacking CREB (CREBαΔ mice) after a 6 min swim stress but not after cocaine exposure (20 mg/kg). Additionally, activation of VTA afferent neurons in the ventral BNST and the infralimbic cortex in CREBαΔ mice was blunted in response to stress. This pattern of neuronal activation persisted in mice that were conditioned to a cocaine place preference procedure before stress exposure. Furthermore, lidocaine inactivation (0.4 µl, 4%) studies demonstrated the necessity of BNST activation for swim-stress-induced reinstatement of cocaine-conditioned reward. Together, the present studies demonstrate that CREB is required for the activation of a unique circuit that converges on the dopamine reward pathway to elicit reinstatement of drug reward and points to the BNST as a key intersection between stress and reward circuits.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Estrés Psicológico , Área Tegmental Ventral/fisiología , Vías Aferentes/efectos de los fármacos , Vías Aferentes/fisiología , Animales , Cocaína/administración & dosificación , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/deficiencia , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Estrés Psicológico/genética , Estrés Psicológico/psicología , Área Tegmental Ventral/efectos de los fármacos
18.
Proc Natl Acad Sci U S A ; 105(30): 10589-94, 2008 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-18645186

RESUMEN

Neural circuits that allow for reciprocal communication between the brain and viscera are critical for coordinating behavior with visceral activity. At the same time, these circuits are positioned to convey signals from pathologic events occurring in viscera to the brain, thereby providing a structural basis for comorbid central and peripheral symptoms. In the pons, Barrington's nucleus and the norepinephrine (NE) nucleus, locus coeruleus (LC), are integral to a circuit that links the pelvic viscera with the forebrain and coordinates pelvic visceral activity with arousal and behavior. Here, we demonstrate that a prevalent bladder dysfunction, produced by partial obstruction in rat, has an enduring disruptive impact on cortical activity through this circuit. Within 2 weeks of partial bladder obstruction, the activity of LC neurons was tonically elevated. LC hyperactivity was associated with cortical electroencephalographic activation that was characterized by decreased low-frequency (1-3 Hz) activity and prominent theta oscillations (6-8 Hz) that persisted for 4 weeks. Selective lesion of the LC-NE system significantly attenuated the cortical effects. The findings underscore the potential for significant neurobehavioral consequences of bladder disorders, including hyperarousal, sleep disturbances, and disruption of sensorimotor integration, as a result of central noradrenergic hyperactivity. The results further imply that pharmacological manipulation of central NE function may alleviate central sequelae of these visceral disorders.


Asunto(s)
Encéfalo/anatomía & histología , Vejiga Urinaria Hiperactiva/etiología , Animales , Encéfalo/patología , Mapeo Encefálico , Electroencefalografía/métodos , Locus Coeruleus/patología , Masculino , Neuronas/metabolismo , Neuronas/patología , Norepinefrina/metabolismo , Puente/patología , Ratas , Ratas Sprague-Dawley , Vejiga Urinaria Hiperactiva/patología , Urodinámica
19.
Neurosci Lett ; 746: 135651, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33482313

RESUMEN

Stress-induced activation of locus coeruleus (LC)-norepinephrine (NE) projections to the prefrontal cortex are thought to promote cognitive responses to stressors. LC activation by stressors is modulated by endogenous opioids that restrain LC activation and facilitate a return to baseline activity upon stress termination. Sex differences in this opioid influence could be a basis for sex differences in stress vulnerability. Consistent with this, we recently demonstrated that µ-opioid receptor (MOR) expression is decreased in the female rat LC compared to the male LC, and this was associated with sexually distinct consequences of activating MOR in the LC on cognitive flexibility. Given that the LC-NE system affects cognitive flexibility through its projections to the medial prefrontal cortex (mPFC), the present study quantified and compared the effects of LC-MOR activation on mPFC neural activity in male and female rats. Local field potential (LFPs) were recorded from the mPFC of freely behaving male and female rats before and following local LC microinjection of the MOR agonist, DAMGO, or vehicle. Intra-LC DAMGO altered the LFP power spectrum selectively in male but not female rats, resulting in a time-dependent increase in the power in delta and alpha frequency bands. LC microinfusion of ACSF had no effect on either sex. Together, the results are consistent with previous evidence for decreased MOR function in the female rat LC and demonstrate that this translates to a diminished effect on cortical activity that can account for sex differences in cognitive consequences. Decreased LC-MOR function in females could contribute to greater stress-induced activation of the LC and increased vulnerability of females to hyperarousal symptoms of stress-related neuropsychiatric pathologies.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Locus Coeruleus/metabolismo , Corteza Prefrontal/metabolismo , Receptores Opioides mu/metabolismo , Caracteres Sexuales , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Femenino , Locus Coeruleus/efectos de los fármacos , Masculino , Microinyecciones/métodos , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/agonistas , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
20.
Front Synaptic Neurosci ; 13: 754786, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34675794

RESUMEN

The pontine nuclei comprising the locus coeruleus (LC) and Barrington's nucleus (BRN) amongst others form the neural circuitry(s) that coordinates arousal and voiding behaviors. However, little is known about the synaptic connectivity of neurons within or across these nuclei. These include corticotropin-releasing factor (CRF+) expressing neurons in the BRN that control bladder contraction and somatostatin expressing (SST+) neurons whose role in this region has not been discerned. To determine the synaptic connectivity of these neurons, we employed optogenetic stimulation with recordings from BRN and LC neurons in brain stem slices of channelrhodopsin-2 expressing SST or CRF neurons. Optogenetic stimulation of CRF+ BRN neurons of Crf Cre ;chr2-yfp mice had little effect on either CRF+ BRN neurons, CRF- BRN neurons, or LC neurons. In contrast, in Sst Cre ;chr2-yfp mice light-activated inhibitory postsynaptic currents (IPSCs) were reliably observed in a majority of LC but not BRN neurons. The GABAA receptor antagonist, bicuculline, completely abolished the light-induced IPSCs. To ascertain if these neurons were part of the neural circuitry that controls the bladder, the trans-synaptic tracer, pseudorabies virus (PRV) was injected into the bladder wall of Crf Cre ;tdTomato or Sst Cre ;tdTomato mice. At 68-72 h post-viral infection, PRV labeled neurons were present only in the BRN, being preponderant in CRF+ neurons with few SST+ BRN neurons labeled from the bladder. At 76 and 96 h post-virus injection, increased labeling was observed in both BRN and LC neurons. Our results suggest SST+ neurons rather than CRF+ neurons in BRN can regulate the activity of LC neurons.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA