Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 27(2): 831-839, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34716408

RESUMEN

Behavioral conditioning and expectation can have profound impact on animal and human physiology. Placebo, administered under positive expectation in clinical trials, can have potent effects on disease pathology, obscuring active medications. Emerging evidence suggests placebo-responsive neurotransmitter systems (e.g., endogenous opioid) regulate immune function by manipulating inflammatory proteins including IL-18, a potent pro-inflammatory, nociceptive cytokine implicated in pathophysiology of various diseases. Validation that neuroimmune interactions involving brain µ-opioid receptor (MOR) activity and plasma IL-18 underlie placebo analgesic expectation could have widespread clinical applications. Unfortunately, current lack of mechanistic clarity obfuscates clinical translation. To elucidate neuroimmune interactions underlying placebo analgesia, we exposed 37 healthy human volunteers to a standardized pain challenge on each of 2 days within a Positron Emission Tomography (PET) neuroimaging paradigm using the MOR selective radiotracer, 11C-Carfentanil (CFN). Each day volunteers received an intervention (placebo under analgesic expectation or no treatment), completed PET scanning, and rated their pain experience. MOR BPND parametric maps were generated from PET scans using standard methods. Results showed placebo reduced plasma IL-18 during pain (W74 = -3.7, p < 0.001), the extent correlating with reduction in pain scores. Placebo reduction in IL-18 covaried with placebo-induced endogenous opioid release in the left nucleus accumbens (T148 = 3.33; puncorr < 0.001) and left amygdala (T148 = 3.30; puncorr < 0.001). These findings are consistent with a modulating effect of placebo (under analgesic expectation in humans) on a potent nociceptive, pro-inflammatory cytokine (IL-18) and underlying relationships with endogenous opioid activity, a neurotransmitter system critically involved in pain, stress, and mood regulation.


Asunto(s)
Analgésicos Opioides , Receptores Opioides mu , Analgésicos , Analgésicos Opioides/metabolismo , Encéfalo/metabolismo , Humanos , Interleucina-18/metabolismo , Neurotransmisores/metabolismo , Péptidos Opioides/metabolismo , Dolor/metabolismo , Tomografía de Emisión de Positrones/métodos , Receptores Opioides mu/metabolismo , Transmisión Sináptica/fisiología
2.
J Neurosci ; 35(27): 9957-65, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26156996

RESUMEN

Back pain is common in the general population, but only a subgroup of back pain patients develops a disabling chronic pain state. The reasons for this are incompletely understood, but recent evidence implies that both preexisting and pain-related variations in the structure and function of the nervous system may contribute significantly to the development of chronic pain. Here, we addressed the role of striatal dopamine (DA) D2/D3 receptor (D2/D3R) function in chronic non-neuropathic back pain (CNBP) by comparing CNBP patients and healthy controls using PET and the D2/D3R-selective radioligand [(11)C]raclopride. D2/D3R availability was measured at baseline and during a pain challenge, yielding in vivo measures of receptor availability (binding potential, BPND) and DA release (change in BPND from baseline to activated state). At baseline, CNBP patients demonstrated reductions in D2/D3R BPND in the ventral striatum compared with controls. These reductions were associated with greater positive affect scores and pain tolerance measures. The reductions in D2/D3R BPND were also correlated with µ-opioid receptor BPND and pain-induced endogenous opioid system activation in the amygdala, further associated with measures of positive affect, the affective component of back pain and pain tolerance. During the pain challenge, lower magnitudes of DA release, and therefore D2/D3R activation, were also found in the ventral striatum in the CNBP sample compared with controls. Our results show that CNBP is associated with adaptations in ventral striatal D2/D3R function, which, together with endogenous opioid system function, contribute to the sensory and affective-motivational features of CNBP. SIGNIFICANCE STATEMENT: The neural systems that underlie chronic pain remain poorly understood. Here, using PET, we provide insight into the molecular mechanisms that regulate sensory and affective dimensions of pain in chronic back pain patients. We found that patients with back pain have alterations in brain dopamine function that are associated with measures of pain sensitivity and affective state, but also with brain endogenous opioid system functional measures. These findings suggest that brain dopamine-opioid interactions are involved in the pathophysiology of chronic pain, which has potential therapeutic implications. Our results may also help to explain individual variation in susceptibility to opioid medication misuse and eventual addiction in the context of chronic pain.


Asunto(s)
Dolor de Espalda/patología , Dopamina/metabolismo , Transmisión Sináptica/fisiología , Estriado Ventral/metabolismo , Estriado Ventral/fisiopatología , Adulto , Analgésicos Opioides/farmacocinética , Mapeo Encefálico , Radioisótopos de Carbono/farmacocinética , Enfermedad Crónica , Antagonistas de Dopamina/farmacocinética , Femenino , Fentanilo/análogos & derivados , Fentanilo/farmacocinética , Humanos , Masculino , Dimensión del Dolor , Unión Proteica/efectos de los fármacos , Racloprida/farmacocinética , Cintigrafía , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/metabolismo , Estriado Ventral/diagnóstico por imagen , Adulto Joven
3.
J Neurosci ; 34(17): 5874-81, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24760847

RESUMEN

Brain-derived neurotrophic factor (BDNF) levels in dopaminergic (DA) cells within the ventral tegmental area (VTA)/nucleus accumbens (NAc) circuitry appear to be a candidate mechanism for the neuroadaptive changes that follow stress and reward responses in animal models. However, the role of the BDNF gene variants in responses to salient cues through DA neurotransmission in humans remains unexplored. Here, we studied the effect of the common functional BDNF Val(66)Met (rs6265) polymorphism on rewarding experiences in the striatum and DA-mediated responses to stress. Seventy-two healthy controls were genotyped for the BDNF Val(66)Met polymorphism and underwent the monetary incentive delay task during an functional magnetic resonance imaging (fMRI) session. Forty-nine of them also underwent a sustained pain challenge with and without placebo administration with potential analgesic properties during PET measures of DA D2/3-receptor-mediated neurotransmission. Neuroimaging results revealed a significant effect of BDNF (Met(66) carriers > Val/Val) on brain responses during the anticipation of monetary losses, baseline D2/3 receptor availability, and pain-stress-induced DA release in the NAc. Conversely, BDNF Met(66) carriers showed no activation in response to monetary gains and a blunted DA response to the analgesic placebo in the NAc. These results provide initial human evidence regarding the effect of the BDNF Val(66)Met polymorphism on DA-mediated responses to stress, its cognitive regulation by positive expectations, and the anticipatory responses to monetary gains and losses in the VTA-NAc pathway. Our results are of relevance to the neurobiology of stress and reward interactions and the pathophysiology of stress-related disorders.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Cuerpo Estriado/fisiología , Neuronas Dopaminérgicas/fisiología , Núcleo Accumbens/fisiología , Polimorfismo de Nucleótido Simple , Recompensa , Estrés Fisiológico/fisiología , Adulto , Mapeo Encefálico , Femenino , Genotipo , Humanos , Imagen por Resonancia Magnética , Masculino , Pruebas Neuropsicológicas , Dolor/fisiopatología , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/genética , Receptores de Dopamina D3/metabolismo , Transmisión Sináptica/fisiología , Área Tegmental Ventral/fisiología
4.
J Neurosci ; 33(37): 14729-37, 2013 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-24027273

RESUMEN

The absence of consistent end organ abnormalities in many chronic pain syndromes has led to a search for maladaptive CNS mechanisms that may explain their clinical presentations and course. Here, we addressed the role of brain regional µ-opioid receptor-mediated neurotransmission, one of the best recognized mechanisms of pain regulation, in chronic back pain in human subjects. We compared µ-opioid receptor availability in vivo at baseline, during pain expectation, and with moderate levels of sustained pain in 16 patients with chronic nonspecific back pain (CNBP) and in 16 age- and gender-matched healthy control subjects, using the µ-opioid receptor-selective radioligand [(11)C]carfentanil and positron emission tomography. We found that CNBP patients showed baseline increases in thalamic µ-opioid receptor availability, contrary to a previously studied sample of patients diagnosed with fibromyalgia. During both pain expectation and sustained pain challenges, CNBP patients showed regional reductions in the capacity to activate this neurotransmitter system compared with their control sample, further associated with clinical pain and affective state ratings. Our results demonstrate heterogeneity in endogenous opioid system functional measures across pain conditions, and alterations in both receptor availability and endogenous opioid function in CNBP that are relevant to the clinical presentation of these patients and the effects of opioid analgesics on µ-opioid receptors.


Asunto(s)
Analgésicos Opioides/metabolismo , Dolor de Espalda/patología , Encéfalo/metabolismo , Adulto , Análisis de Varianza , Dolor de Espalda/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Mapeo Encefálico , Radioisótopos de Carbono , Estudios de Casos y Controles , Enfermedad Crónica , Femenino , Fentanilo/análogos & derivados , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Tomografía de Emisión de Positrones , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
5.
Nature ; 452(7190): 997-1001, 2008 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-18385673

RESUMEN

Understanding inter-individual differences in stress response requires the explanation of genetic influences at multiple phenotypic levels, including complex behaviours and the metabolic responses of brain regions to emotional stimuli. Neuropeptide Y (NPY) is anxiolytic and its release is induced by stress. NPY is abundantly expressed in regions of the limbic system that are implicated in arousal and in the assignment of emotional valences to stimuli and memories. Here we show that haplotype-driven NPY expression predicts brain responses to emotional and stress challenges and also inversely correlates with trait anxiety. NPY haplotypes predicted levels of NPY messenger RNA in post-mortem brain and lymphoblasts, and levels of plasma NPY. Lower haplotype-driven NPY expression predicted higher emotion-induced activation of the amygdala, as well as diminished resiliency as assessed by pain/stress-induced activations of endogenous opioid neurotransmission in various brain regions. A single nucleotide polymorphism (SNP rs16147) located in the promoter region alters NPY expression in vitro and seems to account for more than half of the variation in expression in vivo. These convergent findings are consistent with the function of NPY as an anxiolytic peptide and help to explain inter-individual variation in resiliency to stress, a risk factor for many diseases.


Asunto(s)
Encéfalo/metabolismo , Emociones , Regulación de la Expresión Génica/genética , Variación Genética/genética , Neuropéptido Y/genética , Estrés Fisiológico/genética , Alelos , Ansiedad/genética , Trastornos de Ansiedad/genética , Encéfalo/fisiología , Encéfalo/fisiopatología , Expresión Facial , Finlandia/etnología , Haplotipos/genética , Humanos , Linfocitos/metabolismo , Imagen por Resonancia Magnética , Masculino , Neuropéptido Y/sangre , Péptidos Opioides/metabolismo , Dolor/genética , Polimorfismo de Nucleótido Simple/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estrés Fisiológico/psicología , Estados Unidos/etnología , Población Blanca/genética
6.
J Neurosci ; 32(1): 35-45, 2012 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-22219268

RESUMEN

Functional neuroimaging studies have implicated the prefrontal cortex (PFCTX) in descending modulation of pain and the placebo effect. This study was performed to elucidate comprehensive PFCTX gene expression in an animal model of persistent trigeminal pain. Adult male C57BL/6J mice received facial carrageenan injection and showed sustained increase in nociceptive responses. Microarray analyses of differentially expressed genes in the PFCTX at 3 d after injection showed "immune system process" as the dominant ontology term and increased mRNA expression of S100a8, S100a9, Lcn2, Il2rg, Fcgr1, Fcgr2b, C1qb, Ptprc, Ccl12, and Cd52 were verified by RT-PCR. Upregulation of S100A8, S100A9, and lipocalin 2 (LCN2) were confirmed by Western blots, and cells in the PFCTX were double immunolabeled with MPO, indicating they were neutrophils. Analyses of blood of facial carrageenan-injected mice also showed increased mRNA expression of these markers, suggesting transmigration of activated neutrophils into the brain. Other immune-related genes, Il2rg, Fcgr2b, C1qb, Ptprc, and Ccl12 were upregulated in the PFCTX but not blood. Approximately 70% of S100A9-positive cells in the PFCTX of carrageenan-injected mice were located in capillaries adherent to endothelial cells, whereas 30% were within the brain parenchyma. Carrageenan-injected mice showed significantly reduced nociceptive responses after injection of C terminus of murine S100A9 protein in the lateral ventricles and PFCTX but not somatosensory barrel cortex. Together, these findings demonstrate activation of immune-related genes in the PFCTX during inflammatory pain and highlight an exciting role of neutrophils in linking peripheral inflammation with immune activation of the PFCTX and antinociception.


Asunto(s)
Dolor Crónico/inmunología , Perfilación de la Expresión Génica/métodos , Inhibición Neural/inmunología , Infiltración Neutrófila/inmunología , Corteza Prefrontal/inmunología , Animales , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Dolor Crónico/genética , Dolor Crónico/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibición Neural/genética , Infiltración Neutrófila/genética , Corteza Prefrontal/citología , Corteza Prefrontal/metabolismo
7.
J Neurosci ; 32(44): 15369-76, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23115175

RESUMEN

Neural systems that identify and respond to salient stimuli are critical for survival in a complex and changing environment. In addition, interindividual differences, including genetic variation and hormonal and metabolic status likely influence the behavioral strategies and neuronal responses to environmental challenges. Here, we examined the relationship between leptin allelic variation and plasma leptin levels with DAD2/3R availability in vivo as measured with [(11)C]raclopride PET at baseline and during a standardized pain stress challenge. Allelic variation in the leptin gene was associated with varying levels of dopamine release in response to the pain stressor, but not with baseline D2/3 receptor availability. Circulating leptin was also positively associated with stress-induced dopamine release. These results show that leptin serves as a regulator of neuronal function in humans and provides an etiological mechanism for differences in dopamine neurotransmission in response to salient stimuli as related to metabolic function. The capacity for leptin to influence stress-induced dopaminergic function is of importance for pathological states where dopamine is thought to play an integral role, such as mood, substance-use disorders, eating disorders, and obesity.


Asunto(s)
Dopamina/fisiología , Leptina/fisiología , Estrés Psicológico/fisiopatología , Adulto , Alelos , Índice de Masa Corporal , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Procesamiento de Imagen Asistido por Computador , Leptina/sangre , Leptina/genética , Masculino , Análisis por Micromatrices , Neostriado/diagnóstico por imagen , Neostriado/fisiología , Núcleo Accumbens/diagnóstico por imagen , Núcleo Accumbens/fisiología , Dolor/fisiopatología , Dolor/psicología , Polimorfismo Genético/genética , Polimorfismo de Nucleótido Simple , Tomografía de Emisión de Positrones , Racloprida , Radiofármacos , Adulto Joven
8.
J Neurosci ; 32(27): 9344-50, 2012 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-22764241

RESUMEN

Mesoaccumbal and nigrostriatal projections are sensitive to stress, and heightened stress sensitivity is thought to confer risk for neuropsychiatric disorders. Serotonin 2C (5-HT(2C)) receptors mediate the inhibitory effects of serotonin on dopaminergic circuitry in experimental animals, and preclinical findings have implicated 5-HT(2C) receptors in motivated behaviors and psychotropic drug mechanisms. In humans, a common missense single-nucleotide change (rs6318, Cys23Ser) in the 5-HT(2C) receptor gene (HTR2C) has been associated with altered activity in vitro and with clinical mood disorders. We hypothesized that dopaminergic circuitry would be more sensitive to stress in humans carrying the Ser23 variant. To test this hypothesis, we studied 54 healthy humans using positron emission tomography and the displaceable D(2)/D(3) receptor radiotracer [(11)C]raclopride. Binding potential (BP(ND)) was quantified before and after a standardized stress challenge consisting of 20 min of moderate deep muscular pain, and reduction in BP(ND) served as an index of dopamine release. The Cys23Ser variant was genotyped on a custom array, and ancestry informative markers were used to control for population stratification. We found greater dopamine release in the nucleus accumbens, caudate nucleus, and putamen among Ser23 carriers, after controlling for sex, age, and ancestry. Genotype accounted for 12% of the variance in dopamine release in the nucleus accumbens. There was no association of Cys23Ser with baseline BP(ND). These findings indicate that a putatively functional HTR2C variant (Ser23) is associated with greater striatal dopamine release during pain in healthy humans. Mesoaccumbal stress sensitivity may mediate the effects of HTR2C variation on risk of neuropsychiatric disorders.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Variación Genética/genética , Trastornos del Humor/genética , Polimorfismo Genético/fisiología , Receptor de Serotonina 5-HT2C/genética , Adulto , Cuerpo Estriado/diagnóstico por imagen , Femenino , Humanos , Masculino , Trastornos del Humor/epidemiología , Dolor/epidemiología , Dolor/genética , Cintigrafía , Receptor de Serotonina 5-HT2C/metabolismo , Factores de Riesgo , Adulto Joven
9.
Neuron ; 55(2): 325-36, 2007 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-17640532

RESUMEN

Expectations, positive or negative, are modulating factors influencing behavior. They are also thought to underlie placebo effects, impacting perceptions and biological processes. Using healthy human subjects, we examined the role of the nucleus accumbens (NAC), a region centrally involved in the encoding of reward expectation, in the formation of placebo responses. Employing functional molecular imaging, activation of NAC dopamine (DA) release was observed during placebo administration and related to its anticipated effects, perception-anticipation mismatches, and placebo effect development. In additional functional MRI studies, the expectation of monetary gain increased NAC synaptic activity in a manner proportional to placebo-induced DA release, anticipated effects, perception-anticipation differentials, and actual placebo effects. Individual variations in NAC response to reward expectation accounted for 28% of the variance in the formation of placebo analgesia.


Asunto(s)
Dopamina/metabolismo , Núcleo Accumbens/fisiología , Efecto Placebo , Recompensa , Disposición en Psicología , Adulto , Femenino , Juegos Experimentales , Humanos , Individualidad , Imagen por Resonancia Magnética , Masculino , Núcleo Accumbens/metabolismo , Dolor/metabolismo , Dolor/psicología , Tomografía de Emisión de Positrones , Valores de Referencia
10.
J Pain Res ; 14: 1371-1387, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34079355

RESUMEN

Over the past year our attention has inevitably been on the coronavirus pandemic, the health and welfare of our families, patients, and office staffs as well as the re-opening of our dental practices. In addition, the opioid crisis continues, is very likely to worsen as a result of the pandemic and continues to be a challenge to Dentistry. National public health issues and healthcare disparities continue and have created a global concern for providing evidence-based, adequate pain management in the dental setting. We have brought together a group of national thought leaders and experts in this field who will share their insights on the current state of opioid prescribing in Dentistry and describe some of the exciting work being done in advancing pain management. The learning objectives for this conference proceedings were: Describing the implications of current public health concerns for safe and effective pain management in dental medicine.Identifying risk factors and understanding the current guidelines for the use of opioid and non-opioid medications in dental medicine.Analyzing the interprofessional collaborations necessary for effective pain management in dental medicine.Recognizing the challenges and opportunities brought about by the COVID-19 pandemic for the dental profession.Applying evidence-based strategies for managing the complex pain patient in the dental setting.Appraising new and future modalities for the assessment and management of orofacial pain.

11.
Mol Neurobiol ; 56(2): 1137-1166, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29876878

RESUMEN

The prefrontal cortex (PFC) is not only important in executive functions, but also pain processing. The latter is dependent on its connections to other areas of the cerebral neocortex, hippocampus, periaqueductal gray (PAG), thalamus, amygdala, and basal nuclei. Changes in neurotransmitters, gene expression, glial cells, and neuroinflammation occur in the PFC during acute and chronic pain, that result in alterations to its structure, activity, and connectivity. The medial PFC (mPFC) could serve dual, opposing roles in pain: (1) it mediates antinociceptive effects, due to its connections with other cortical areas, and as the main source of cortical afferents to the PAG for modulation of pain. This is a 'loop' where, on one side, a sensory stimulus is transformed into a perceptual signal through high brain processing activity, and perceptual activity is then utilized to control the flow of afferent sensory stimuli at their entrance (dorsal horn) to the CNS. (2) It could induce pain chronification via its corticostriatal projection, possibly depending on the level of dopamine receptor activation (or lack of) in the ventral tegmental area-nucleus accumbens reward pathway. The PFC is involved in biopsychosocial pain management. This includes repetitive transcranial magnetic stimulation, transcranial direct current stimulation, antidepressants, acupuncture, cognitive behavioral therapy, mindfulness, music, exercise, partner support, empathy, meditation, and prayer. Studies demonstrate the role of the PFC during placebo analgesia, and in establishing links between pain and depression, anxiety, and loss of cognition. In particular, losses in PFC grey matter are often reversible after successful treatment of chronic pain.


Asunto(s)
Percepción del Dolor/fisiología , Dolor/fisiopatología , Corteza Prefrontal/fisiopatología , Animales , Dopamina/metabolismo , Humanos , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Dolor/metabolismo , Corteza Prefrontal/metabolismo
12.
Nihon Hotetsu Shika Gakkai Zasshi ; 52(2): 200-4, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18467792

RESUMEN

PURPOSE: To clarify the section showing minimal intraindividual variations in the movement of the mandibular incisal point during mastication of softened chewing gum. METHODS: Twenty healthy subjects were asked to chew softened chewing gum on the habitual side for 20 seconds. The change in the spatial parameters (gape and masticatory width) and temporal parameter (cycle time) were investigated for 20 cycles from the first cycle. The coefficients of variation of these parameters were investigated for each of 10 consecutive cycles (first to eleventh series). RESULTS: The spatial and temporal parameters were maximal at the first cycle, decreased progressively until the fourth or fifth cycle, and then remained almost unchanged thereafter. The coefficients of variation of the parameters were maximal during the first series, decreased progressively until the fourth to sixth series, and then tended to increase gradually thereafter. Minimal coefficients of variation were observed during the fifth and sixth series for the gape, during the fifth series for the width, and during the fourth series for the cycle time. CONCLUSION: These results suggest that the ten cycles after the fourth to the sixth cycle was the section showing minimal intra-individual variations in the masticatory movement during the chewing of softened chewing gum.


Asunto(s)
Individualidad , Masticación/fisiología , Sistema Estomatognático/fisiología , Adulto , Femenino , Humanos , Masculino , Mandíbula/fisiología
13.
J Neurosci ; 26(42): 10789-95, 2006 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-17050717

RESUMEN

In addition to its involvement in motor control and in encoding reward value, increasing evidence also implicates basal ganglia dopaminergic mechanisms in responses to stress and aversive stimuli. Basal ganglia dopamine (DA) neurotransmission may then respond to environmental events depending on their saliency, orienting the subsequent responses of the organism to both positive and negative stimuli. Here we examined the involvement of DA neurotransmission in the human response to pain, a robust physical and emotional stressor across species. Positron emission tomography with the DA D2 receptor antagonist radiotracer [11C]raclopride detected significant activation of DA release in dorsal and ventral regions of the basal ganglia of healthy volunteers. Activation of nigrostriatal (dorsal nucleus caudate and putamen) DA D2 receptor-mediated neurotransmission was positively associated with individual variations in subjective ratings of sensory and affective qualities of the pain. In contrast, mesolimbic (nucleus accumbens) DA activation, which may impact on both D2 and D3 receptors, was exclusively associated with variations in the emotional responses of the individual during the pain challenge (increases in negative affect and fear ratings). These data demonstrate that basal ganglia dopamine D2 receptor-mediated neurotransmission is involved in responses to pain and that it contributes to individual variations in the pain experience at the levels of physical and emotional elements, albeit with different neuroanatomical substrates.


Asunto(s)
Ganglios Basales/metabolismo , Dopamina/metabolismo , Dimensión del Dolor/métodos , Dolor/metabolismo , Estrés Fisiológico/metabolismo , Adulto , Ganglios Basales/efectos de los fármacos , Antagonistas de Dopamina/farmacología , Antagonistas de los Receptores de Dopamina D2 , Femenino , Humanos , Masculino , Dolor/psicología , Dimensión del Dolor/efectos de los fármacos , Tomografía de Emisión de Positrones/métodos , Receptores de Dopamina D2/metabolismo , Estrés Fisiológico/psicología
14.
J Neurosci ; 26(21): 5777-85, 2006 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-16723535

RESUMEN

Prominent interindividual and sex-dependent differences have been described in responses to sustained pain and other stressful stimuli. Variations in mu-opioid receptor-mediated endogenous opioid neurotransmission may underlie some of these processes. We examined both baseline mu-opioid receptor levels and the activation of this neurotransmitter system during sustained pain using positron emission tomography in a sample of young healthy men and women. Women were studied twice, during low and high estrogen states. The high-estrogen state was associated with regional increases in baseline mu-opioid receptor availability in vivo and a greater activation of endogenous opioid neurotransmission during the pain stressor. The latter did not differ from that obtained in males. During the low estrogen condition, however, significant reductions in endogenous opioid tone were observed at the level of thalamus, nucleus accumbens, and amygdala, which were associated with hyperalgesic responses. Estrogen-associated variations in the activity of mu-opioid neurotransmission correlated with individual ratings of the sensory and affective perceptions of the pain and the subsequent recall of that experience. These data demonstrate a significant role of estrogen in modulating endogenous opioid neurotransmission and associated psychophysical responses to a pain stressor in humans.


Asunto(s)
Analgésicos/metabolismo , Encéfalo/metabolismo , Estradiol/sangre , Narcóticos/metabolismo , Dolor/metabolismo , Receptores Opioides mu/metabolismo , Transmisión Sináptica/efectos de los fármacos , Adulto , Femenino , Humanos , Masculino , Factores Sexuales
16.
J Neurosci ; 25(34): 7754-62, 2005 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-16120776

RESUMEN

Reductions in pain ratings when administered a placebo with expected analgesic properties have been described and hypothesized to be mediated by the pain-suppressive endogenous opioid system. Using molecular imaging techniques, we directly examined the activity of the endogenous opioid system on mu-opioid receptors in humans in sustained pain with and without the administration of a placebo. Significant placebo-induced activation of mu-opioid receptor-mediated neurotransmission was observed in both higher-order and sub-cortical brain regions, which included the pregenual and subgenual rostral anterior cingulate, the dorsolateral prefrontal cortex, the insular cortex, and the nucleus accumbens. Regional activations were paralleled by lower ratings of pain intensity, reductions in its sensory and affective qualities, and in the negative emotional state of the volunteers. These data demonstrate that cognitive factors (e.g., expectation of pain relief) are capable of modulating physical and emotional states through the site-specific activation of mu-opioid receptor signaling in the human brain.


Asunto(s)
Péptidos Opioides/fisiología , Dimensión del Dolor/métodos , Receptores Opioides mu/fisiología , Adulto , Analgésicos Opioides/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Humanos , Masculino , Naloxona/farmacología , Péptidos Opioides/metabolismo , Dimensión del Dolor/efectos de los fármacos , Efecto Placebo , Tomografía de Emisión de Positrones/métodos , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/metabolismo
17.
J Neurosci ; 22(12): 5100-7, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12077205

RESUMEN

Sex differences in the experience of clinical and experimental pain have been reported. However, the neurobiological sources underlying the variability in pain responses between sexes have not been adequately explored, especially in humans. The endogenous opioid neurotransmitters and mu-opioid receptors are centrally implicated in responses to stress, in the suppression of pain, and in the action of opiate analgesic drugs. Here we examined sex differences in the activation of the mu-opioid system in response to an intensity-controlled sustained deep-tissue pain challenge with positron emission tomography and a mu-opioid receptor-selective radiotracer. Twenty-eight young healthy volunteers (14 men and 14 women) were studied during saline control and pain conditions using a double-blind, randomized, and counterbalanced design. Women were scanned during the early follicular phase of their menstrual cycles after ovulatory cycles. Significant sex differences in the regional activation of the mu-opioid system in response to sustained pain were detected compared with saline controls. Men demonstrated larger magnitudes of mu-opioid system activation than women in the anterior thalamus, ventral basal ganglia, and amygdala. Conversely, women demonstrated reductions in the basal state of activation of the mu-opioid system during pain in the nucleus accumbens, an area previously associated with hyperalgesic responses to the blockade of opioid receptors in experimental animals. These data demonstrate that at matched levels of pain intensity, men and women during their follicular phase differ in the magnitude and direction of response of the mu-opioid system in distinct brain nuclei.


Asunto(s)
Dolor/etiología , Receptores Opioides mu/fisiología , Factores Sexuales , Adulto , Amígdala del Cerebelo/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Método Doble Ciego , Femenino , Humanos , Masculino , Núcleo Accumbens/diagnóstico por imagen , Dolor/diagnóstico por imagen , Tálamo/diagnóstico por imagen , Tomografía Computarizada de Emisión
18.
Artículo en Inglés | MEDLINE | ID: mdl-16256255

RESUMEN

Unraveling the pathways and neurobiological mechanisms that underlie the regulation of physical and emotional stress responses in humans is of critical importance to understand vulnerability and resiliency factors to the development of a number of complex physical and psychopathological states. Dysregulation of central stress response circuits have been implicated in the establishment of conditions as diverse as persistent pain, mood and personality disorders and substance abuse and dependence. The present review examines the contribution of the endogenous opioid system and mu-opioid receptors to the modulation and adaptation of the organism to challenges, such as sustained pain and negative emotional states, which threaten its internal homeostasis. Data accumulated in animal models, and more recently in humans, point to this neurotransmitter system as a critical modulator of the transition from acute (warning signals) to sustained (stressor) environmental adversity. The existence of pathways and regulatory mechanisms common to the regulation of both physical and emotional states transcend classical categorical disease classifications, and point to the need to utilize dimensional, "symptom"-related approximations to their study. Possible future areas of study at the interface of "mind" (cognitive-emotional) and "body" (physical) functions are delineated in this context.


Asunto(s)
Emociones/fisiología , Endorfinas/fisiología , Receptores Opioides mu/fisiología , Estrés Psicológico/fisiopatología , Animales , Femenino , Humanos , Dolor/fisiopatología , Tomografía de Emisión de Positrones , Embarazo , Estrés Psicológico/diagnóstico por imagen
19.
J Can Dent Assoc ; 71(5): 332, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15949256

RESUMEN

There is considerable concern among leaders in both academia and the prosthodontic profession about the vitality of prosthodontic research and the discipline in general. Many feel that prosthodontics should be focused more on issues of societal significance. In addition, patient-oriented research is becoming more difficult to support within the current climate of ever-lower priority for discipline-based research.A break with traditional lines of enquiry is required, which will entail a corresponding break with established departmental boundaries, to gain access to a multiplicity of complementary skills. Several themes will be crucial in future prosthodontic research: clinical decision making, including health economics; materials science and host response at the implant interface; biocompatibility, functional properties and serviceability of prosthodontic materials; and function and dysfunction of the masticatory system. These themes are at the core of future projects that will address quality-of-life issues related to tooth loss. Prosthodontic researchers will have to be far more aggressive in developing synergistic collaborative arrangements, to align prosthodontic research with the major issues of the day, such as aging of the population, health disparities and access to preventive strategies. Through these collaborations, prosthodontics will remain a flagship discipline within dentistry, and its practitioners will be engaged in the major issues of health care.


Asunto(s)
Investigación Dental/tendencias , Prostodoncia/tendencias , Humanos , Relaciones Interprofesionales , Apoyo a la Investigación como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA