Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
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.

2.
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.

3.
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
4.
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
5.
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
6.
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
7.
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
11.
Neurobiol Stress ; 10: 100165, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31193524

RESUMEN

Chronic stress can lead to psychiatric illness characterized by impairments of executive function, implicating the prefrontal cortex as a target of stress-related pathology. Previous studies have shown that various types of chronic stress paradigms reduce dendritic branching, length and spines of medial prefrontal cortex (mPFC) pyramidal neurons. However, these studies largely focused on layer II/III pyramidal neurons in adult male rats with less known about layer V, the site of projection neurons. Because the prefrontal cortex develops throughout adolescence, stress during adolescence may have a greater impact on structure and function than stress occurring during adulthood. Furthermore, females display greater risk of stress-related psychiatric disorders, indicating sex-specific responses to stress. In this study, male and female adolescent (42-48 days old, 4 rats per group) or adult (68-72 days old, 4 rats per group) Sprague-Dawley rats were exposed to 5 days of repeated social stress in the resident-intruder paradigm or control manipulation. We examined dendritic morphology of cells in the mPFC in both layer II/III and Layer V. Repeated social stress resulted in decreased dendritic branching in layer II/III apical dendrites regardless of sex or age. In apical layer V dendrites, stress increased branching in adult males but decreased it in all other groups. Stress resulted in a decrease in dendritic spines in layer V apical dendrites for male adolescents and female adults, and this was mostly due to a decrease in filopodial and mushroom spines for male adolescents, but stubby spines for female adults. In sum, these results suggest that repeated stress reduces complexity and synaptic connectivity in adolescents and female adults in both input and output layers of prelimbic mPFC, but not in male adults. These changes may represent a potential underlying mechanism as to why adolescents and females are more susceptible to the negative cognitive effects of repeated or chronic stress.

12.
Neurosci Biobehav Rev ; 103: 50-59, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31212019

RESUMEN

The neuropeptide, corticotropin-releasing factor (CRF), is a key modulator of physiological, endocrine, and behavioral responses during stress. Dysfunction of the CRF system has been observed in stress-related affective disorders including post-traumatic stress disorder, depression, and anxiety. Beyond affective symptoms, these disorders are also characterized by impaired cognition, for which current pharmacological treatments are lacking. Thus, there is a need for pro-cognitive treatments to improve quality of life for individuals suffering from mental illness. In this review, we highlight research demonstrating that CRF elicits potent modulatory effects on higher-order cognition via actions within the prefrontal cortex and subcortical monoaminergic and cholinergic systems. Additionally, we identify questions for future preclinical research on this topic, such as the need to investigate sex differences in the cognitive and microcircuit actions of CRF, and whether CRF may represent a pharmacological target to treat cognitive dysfunction. Addressing these questions will provide new insight into pathophysiology underlying cognitive dysfunction and may lead to improved treatments for neuropsychiatric disorders.


Asunto(s)
Atención/fisiología , Encéfalo/fisiología , Cognición/fisiología , Disfunción Cognitiva , Hormona Liberadora de Corticotropina/fisiología , Toma de Decisiones/fisiología , Memoria a Corto Plazo/fisiología , Motivación/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Humanos , Masculino
14.
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
15.
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
16.
Neuropsychopharmacology ; 43(13): 2514-2520, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30250308

RESUMEN

Mu opioid receptor agonists are among the most powerful analgesic medications but also among the most addictive. The current opioid crisis has energized a quest to develop opioid analgesics that are devoid of untoward effects. Since their discovery in the 1970's, there have been major advances in our understanding of the endogenous opioid systems that these drugs target. Yet many questions remain and the development of non-addictive opioid analgesics has not been achieved. However, access to new molecular, genetic and computational tools have begun to elucidate the structural dynamics of opioid receptors, the scaffolding that links them to intracellular signaling cascades, their cellular trafficking and the distinct ways that various opioid drugs modify them. This mini-review highlights some of the chemical and pharmacological findings and new perspectives that have arisen from studies using these tools. They reveal multiple layers of complexity of opioid receptor function, including a spatiotemporal specificity in opioid receptor-induced cellular signaling, ligand-directed biased signaling, allosteric modulation of ligand interactions, heterodimerization of different opioid receptors, and the existence of slice variants with different ligand specificity. By untangling these layers, basic research into the chemistry and pharmacology of opioid receptors is guiding the way towards deciphering the mysteries of tolerance and physical dependence that have plagued the field and is providing a platform for the development of more effective and safer opioids.


Asunto(s)
Analgésicos Opioides/metabolismo , Encéfalo/metabolismo , Receptores Opioides/fisiología , Afecto/efectos de los fármacos , Afecto/fisiología , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Humanos , Dolor/tratamiento farmacológico , Dolor/metabolismo , Receptores Opioides/agonistas , Receptores Opioides/química , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
17.
Elife ; 62017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29199948

RESUMEN

Urodynamic status must interact with arousal and attentional processes so that voiding occurs under appropriate conditions. To elucidate the central encoding of this visceral demand, multisite recordings were made within a putative pontine-cortical micturition circuit from the pontine micturition center (PMC), locus coeruleus (LC) and medial prefrontal cortex (mPFC) during cystometry in unanesthetized rats. PMC neurons had homogeneous firing patterns, characterized by tonic activity and phasic bursts that were temporally associated with distinct phases of the micturition cycle. LC and cortical activation became synchronized 20-30 s prior to micturition. During this pre-micturition interval, a theta oscillation developed in the LC, the mPFC desynchronized and LC-mPFC coherence increased in the theta frequency range. The temporal offset between the shift in LC-mPFC network activity and micturition may allow time to disengage from ongoing behaviors unrelated to micturition and initiate specific voiding behaviors so that micturition occurs in environmentally and socially appropriate conditions.


Asunto(s)
Tronco Encefálico/fisiología , Red Nerviosa/fisiología , Vejiga Urinaria/fisiología , Micción , Potenciales de Acción , Animales , Electroencefalografía , Femenino , Ratas Sprague-Dawley
18.
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
19.
Neurosci Biobehav Rev ; 74(Pt B): 393-400, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27131968

RESUMEN

Chronic exposure to psychosocial stress has adverse effects on cardiovascular health, however the stress-sensitive neurocircuitry involved remains to be elucidated. The anatomical and physiological characteristics of the locus coeruleus (LC)-norepinephrine (NE) system position it to contribute to stress-induced cardiovascular disease. This review focuses on cardiovascular dysfunction produced by social stress and a major theme highlighted is that differences in coping strategy determine individual differences in social stress-induced cardiovascular vulnerability. The establishment of different coping strategies and cardiovascular vulnerability during repeated social stress has recently been shown to parallel a unique plasticity in LC afferent regulation, resulting in either excitatory or inhibitory input to the LC. This contrasting regulation of the LC would translate to differences in cardiovascular regulation and may serve as the basis for individual differences in the cardiopathological consequences of social stress. The advances described suggest new directions for developing treatments and/or strategies for decreasing stress-induced cardiovascular vulnerability.


Asunto(s)
Enfermedades Cardiovasculares , Estrés Psicológico , Adaptación Psicológica , Encéfalo , Humanos , Locus Coeruleus , Norepinefrina
20.
Physiol Behav ; 172: 40-48, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27423323

RESUMEN

Repeated exposure to psychosocial stress is a robust sympathomimetic stressor and as such has adverse effects on cardiovascular health. While the neurocircuitry involved remains unclear, the physiological and anatomical characteristics of the locus coeruleus (LC)-norepinephrine (NE) system suggest that it is poised to contribute to stress-induced cardiovascular vulnerability. A major theme throughout is to review studies that shed light on the role that the LC may play in individual differences in vulnerability to social stress-induced cardiovascular dysfunction. Recent findings are discussed that support a unique plasticity in afferent regulation of the LC, resulting in either excitatory or inhibitory input to the LC during establishment of different stress coping strategies. This contrasting regulation of the LC by either afferent regulation, or distinct differences in stress-induced neuroinflammation would translate to differences in cardiovascular regulation and may serve as the basis for individual differences in the cardiopathological consequences of social stress. The goal of this review is to highlight recent developments in the interplay between the LC-NE and cardiovascular systems during repeated stress in an effort to advance therapeutic treatments for the development of stress-induced cardiovascular vulnerability.


Asunto(s)
Sistema Cardiovascular/fisiopatología , Individualidad , Locus Coeruleus/fisiología , Locus Coeruleus/fisiopatología , Norepinefrina/fisiología , Estrés Psicológico/fisiopatología , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...