Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 587(7833): 258-263, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33116307

RESUMEN

The anterolateral pathway consists of ascending spinal tracts that convey pain, temperature and touch information from the spinal cord to the brain1-4. Projection neurons of the anterolateral pathway are attractive therapeutic targets for pain treatment because nociceptive signals emanating from the periphery are channelled through these spinal projection neurons en route to the brain. However, the organizational logic of the anterolateral pathway remains poorly understood. Here we show that two populations of projection neurons that express the structurally related G-protein-coupled receptors (GPCRs) TACR1 and GPR83 form parallel ascending circuit modules that cooperate to convey thermal, tactile and noxious cutaneous signals from the spinal cord to the lateral parabrachial nucleus of the pons. Within this nucleus, axons of spinoparabrachial (SPB) neurons that express Tacr1 or Gpr83 innervate distinct sets of subnuclei, and strong optogenetic stimulation of the axon terminals induces distinct escape behaviours and autonomic responses. Moreover, SPB neurons that  express Gpr83 are highly sensitive to cutaneous mechanical stimuli and receive strong synaptic inputs from both high- and low-threshold primary mechanosensory neurons. Notably, the valence associated with activation of SPB neurons that express Gpr83 can be either positive or negative, depending on stimulus intensity. These findings reveal anatomically, physiologically and functionally distinct subdivisions of the SPB tract that underlie affective aspects of touch and pain.


Asunto(s)
Vías Nerviosas , Dolor/fisiopatología , Médula Espinal/citología , Médula Espinal/fisiología , Tacto/fisiología , Animales , Axones/metabolismo , Femenino , Masculino , Mecanotransducción Celular , Ratones , Filosofía , Receptores Acoplados a Proteínas G/genética , Células Receptoras Sensoriales/metabolismo , Piel/inervación , Sinapsis/metabolismo
2.
J Neurosci ; 42(43): 8095-8112, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36104281

RESUMEN

Intracortical inhibition in motor cortex (M1) regulates movement and motor learning. If cortical and thalamic inputs target different inhibitory cell types in different layers, then these afferents may play different roles in regulating M1 output. Using mice of both sexes, we quantified input to two main classes of M1 interneurons, parvalbumin+ (PV+) cells and somatostatin+ (SOM+) cells, using monosynaptic rabies tracing. We then compared anatomic and functional connectivity based on synaptic strength from sensory cortex and thalamus. Functionally, each input innervated M1 interneurons with a unique laminar profile. Different interneuron types were excited in a distinct, complementary manner, suggesting feedforward inhibition proceeds selectively via distinct circuits. Specifically, somatosensory cortex (S1) inputs primarily targeted PV+ neurons in upper layers (L2/3) but SOM+ neurons in middle layers (L5). Somatosensory thalamus [posterior nucleus (PO)] inputs targeted PV+ neurons in middle layers (L5). In contrast to sensory cortical areas, thalamic input to SOM+ neurons was equivalent to that of PV+ neurons. Thus, long-range excitatory inputs target inhibitory neurons in an area and a cell type-specific manner, which contrasts with input to neighboring pyramidal cells. In contrast to feedforward inhibition providing generic inhibitory tone in cortex, circuits are selectively organized to recruit inhibition matched to incoming excitatory circuits.SIGNIFICANCE STATEMENT M1 integrates sensory information and frontal cortical inputs to plan and control movements. Although inputs to excitatory cells are described, the synaptic circuits by which these inputs drive specific types of M1 interneurons are unknown. Anatomical results with rabies tracing and physiological quantification of synaptic strength shows that two main classes of inhibitory cells (PV+ and SOM+ interneurons) both receive substantial cortical and thalamic input, in contrast to interneurons in sensory areas (where thalamic input strongly prefers PV+ interneurons). Further, each input studied targets PV+ and SOM+ interneurons in a different fashion, suggesting that separate, specific circuits exist for recruitment of feedforward inhibition.


Asunto(s)
Corteza Motora , Rabia , Femenino , Masculino , Ratones , Animales , Parvalbúminas/metabolismo , Corteza Motora/metabolismo , Rabia/metabolismo , Tálamo/fisiología , Neuronas/fisiología , Interneuronas/fisiología , Somatostatina/metabolismo
3.
Anesthesiology ; 139(4): 462-475, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37364291

RESUMEN

BACKGROUND: Pharmacologic manipulations directed at the periaqueductal gray have demonstrated the importance of the µ-opioid receptor in modulating reflexive responses to nociception. The authors hypothesized that a supraspinal pathway centered on neurons in the periaqueductal gray containing the µ-opioid receptor could modulate nociceptive and itch behaviors. METHODS: The study used anatomical, optogenetic, and chemogenetic approaches in male and female mice to manipulate µ-opioid receptor neurons in the periaqueductal gray. Behavioral assays including von Frey, Hargreaves, cold plantar, chloroquine-induced itch, hotplate, formalin-induced injury, capsaicin-induced injury, and open field tests were used. In separate experiments, naloxone was administered in a postsurgical model of latent sensitization. RESULTS: Activation of µ-opioid receptor neurons in the periaqueductal gray increased jumping (least-squares mean difference of -3.30 s; 95% CI, -6.17 to -0.44; P = 0.023; n = 7 or 8 mice per group), reduced itch responses (least-squares mean difference of 70 scratching bouts; 95% CI, 35 to 105; P < 0.001; n = 8 mice), and elicited modestly antinociceptive effects (least-squares mean difference of -0.7 g on mechanical and -10.24 s on thermal testing; 95% CI, -1.3 to -0.2 and 95% CI, -13.77 to -6.70, and P = 0.005 and P < 0.001, respectively; n = 8 mice). Last, the study uncovered the role of the periaqueductal gray in suppressing hyperalgesia after a postsurgical state of latent sensitization (least-squares mean difference comparing saline and naloxone of -12 jumps; 95% CI, -17 to -7; P < 0.001 for controls; and -2 jumps; 95% CI, -7 to 4; P = 0.706 after optogenetic stimulation; n = 7 to 9 mice per group). CONCLUSIONS: µ-Opioid receptor neurons in the periaqueductal gray modulate distinct nocifensive behaviors: their activation reduced responses to mechanical and thermal testing, and attenuated scratching behaviors, but facilitated escape responses. The findings emphasize the role of the periaqueductal gray in the behavioral expression of nociception using reflexive and noxious paradigms.


Asunto(s)
Nocicepción , Sustancia Gris Periacueductal , Ratones , Masculino , Femenino , Animales , Sustancia Gris Periacueductal/fisiología , Naloxona/farmacología , Neuronas/metabolismo , Receptores Opioides , Receptores Opioides mu/fisiología
4.
Brain ; 145(7): 2586-2601, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35598161

RESUMEN

In perilous and stressful situations, the ability to suppress pain can be critical for survival. The rostral ventromedial medulla contains neurons that robustly inhibit nocioception at the level of the spinal cord through a top-down modulatory pathway. Although much is known about the role of the rostral ventromedial medulla in the inhibition of pain, the precise ability to directly manipulate pain-inhibitory neurons in the rostral ventromedial medulla has never been achieved. We now expose a cellular circuit that inhibits nocioception and itch in mice. Through a combination of molecular, tracing and behavioural approaches, we found that rostral ventromedial medulla neurons containing the kappa-opioid receptor inhibit itch and nocioception. With chemogenetic inhibition, we uncovered that these neurons are required for stress-induced analgesia. Using intersectional chemogenetic and pharmacological approaches, we determined that rostral ventromedial medulla kappa-opioid receptor neurons inhibit nocioception and itch through a descending circuit. Lastly, we identified a dynorphinergic pathway arising from the periaqueductal grey that modulates nociception within the rostral ventromedial medulla. These discoveries highlight a distinct population of rostral ventromedial medulla neurons capable of broadly and robustly inhibiting itch and nocioception.


Asunto(s)
Bulbo Raquídeo , Neuronas , Dolor , Prurito , Receptores Opioides kappa , Animales , Bulbo Raquídeo/citología , Ratones , Neuronas/fisiología , Dolor/fisiopatología , Prurito/fisiopatología , Receptores Opioides kappa/metabolismo
5.
J Neurosci ; 40(46): 8816-8830, 2020 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-33051347

RESUMEN

The neurokinin-1 receptor (NK1R; encoded by Tacr1) is expressed in spinal dorsal horn neurons and has been suggested to mediate itch in rodents. However, previous studies relied heavily on neurotoxic ablation of NK1R spinal neurons, which limited further dissection of their function in spinal itch circuitry. To address this limitation, we leveraged a newly developed Tacr1CreER mouse line to characterize the role of NK1R spinal neurons in itch. We show that pharmacological activation of spinal NK1R and chemogenetic activation of Tacr1CreER spinal neurons increases itch behavior in male and female mice, whereas pharmacological inhibition of spinal NK1R suppresses itch behavior. We use fluorescence in situ hybridization (FISH) to characterize the endogenous expression of Tacr1 throughout the superficial and deeper dorsal horn (DDH), as well as the lateral spinal nucleus (LSN), of mouse and human spinal cord. Retrograde labeling studies in mice from the parabrachial nucleus (PBN) show that less than 20% of superficial Tacr1CreER dorsal horn neurons are spinal projection neurons, and thus the majority of Tacr1CreER are local interneurons. We then use a combination of in situ hybridization and ex vivo two-photon Ca2+ imaging of the mouse spinal cord to establish that NK1R and the gastrin-releasing peptide receptor (GRPR) are coexpressed within a subpopulation of excitatory superficial dorsal horn (SDH) neurons. These findings are the first to suggest a role for NK1R interneurons in itch and extend our understanding of the complexities of spinal itch circuitry.SIGNIFICANCE STATEMENT The spinal cord is a critical hub for processing somatosensory input, yet which spinal neurons process itch input and how itch signals are encoded within the spinal cord is not fully understood. We demonstrate neurokinin-1 receptor (NK1R) spinal neurons mediate itch behavior in mice and that the majority of NK1R spinal neurons are local interneurons. These NK1R neurons comprise a subset of gastrin-releasing peptide receptor (GRPR) interneurons and are thus positioned at the center of spinal itch transmission. We show NK1R mRNA expression in human spinal cord, underscoring the translational relevance of our findings in mice. This work is the first to suggest a role for NK1R interneurons in itch and extends our understanding of the complexities of spinal itch circuitry.


Asunto(s)
Interneuronas , Red Nerviosa/fisiopatología , Prurito/fisiopatología , Receptores de Bombesina/biosíntesis , Receptores de Bombesina/genética , Receptores de Neuroquinina-1/biosíntesis , Receptores de Neuroquinina-1/genética , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Adulto , Animales , Conducta Animal , Plexo Braquial/fisiopatología , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Dolor/psicología , Células del Asta Posterior/metabolismo , Prurito/genética , Prurito/psicología
6.
Anesthesiology ; 135(2): 350-365, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34237130

RESUMEN

Opioids are a mainstay of treatment for pain worldwide. Pruritus, a common side effect of opioids, is a patient dissatisfier that limits their use in many clinical settings. Both parenteral and neuraxial administration of opioids frequently evoke pruritus. The ability of opioids to suppress pain while causing itch continues to perplex clinicians and researchers alike. Several mechanisms have been proposed to explain how opioids can give rise to pruritus, but specific knowledge gaps perpetuate debate. This review summarizes the clinical burden of opioid-induced pruritus and emphasizes recent discoveries of peripheral and central mechanisms for opioid-induced pruritus, particularly with respect to scientific and conceptual advances in spinal cord circuitry and mast cell biology. The mechanisms and effectiveness of existing medications used for clinical management of pruritus will be evaluated, and we will highlight the emerging preclinical utility of selective κ-opioid receptor agonists, such as nalfurafine, for the management of opioid-induced pruritus.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Prurito/inducido químicamente , Prurito/terapia , Humanos , Infusiones Parenterales , Infusión Espinal
7.
Proc Natl Acad Sci U S A ; 112(23): E3010-9, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26023183

RESUMEN

Retinal bipolar (BP) cells mediate the earliest steps in image processing in the visual system, but the genetic pathways that regulate their development and function are incompletely known. We identified PRDI-BF1 and RIZ homology domain containing 8 (PRDM8) as a highly conserved transcription factor that is abundantly expressed in mouse retina. During development and in maturity, PRDM8 is expressed strongly in BP cells and a fraction of amacrine and ganglion cells. To determine whether Prdm8 is essential to BP cell development or physiology, we targeted the gene in mice. Prdm8(EGFP/EGFP) mice showed nonprogressive b-wave deficits on electroretinograms, consistent with compromised BP cell function or circuitry resembling the incomplete form of human congenital stationary night blindness (CSNB). BP cell specification was normal in Prdm8(EGFP/EGFP) retina as determined by VSX2(+) cell numbers and retinal morphology at postnatal day 6. BP subtype differentiation was impaired, however, as indicated by absent or diminished expression of BP subtype-specific markers, including the putative PRDM8 regulatory target PKCα (Prkca) and its protein. By adulthood, rod bipolar (RB) and type 2 OFF-cone bipolar (CB) cells were nearly absent from Prdm8-null mice. Although no change was detected in total amacrine cell (AC) numbers, increased PRKCA(+) and cholinergic ACs and decreased GABAergic ACs were seen, suggesting an alteration in amacrine subtype identity. These findings establish that PRDM8 is required for RB and type 2 OFF-CB cell survival and amacrine subtype identity, and they present PRDM8 as a candidate gene for human CSNB.


Asunto(s)
Células Amacrinas/citología , Supervivencia Celular/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Células Bipolares de la Retina/citología , Células Amacrinas/metabolismo , Animales , Proteínas de Unión al ADN , Histona Metiltransferasas , Ratones , Ratones Transgénicos , Células Bipolares de la Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/citología , Células Fotorreceptoras Retinianas Bastones/metabolismo
8.
Dysphagia ; 33(6): 739-748, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29619560

RESUMEN

This study prospectively evaluated relationships between oral morbidities and swallowing ability in head/neck cancer patients following chemoradiation therapy (CRT) and at 3 months following CRT. Thirty patients with confirmed head/neck cancer undergoing chemoradiation were assessed with a battery of swallowing measures and measures of oral morbidities related to chemoradiation (xerostomia, mucositis, pain, taste/smell, oral moisture). All measures were completed at baseline (within the first week of CRT), at 6 weeks (end of treatment), and at 3 months following chemoradiation. Descriptive and univariate statistics were used to depict change over time in swallowing and each oral morbidity. Correlation analyses evaluated relationships between swallowing function and oral morbidities at each time point. Most measures demonstrated significant negative change at 6 weeks with incomplete recovery at 3 months. At 6 weeks, mucositis ratings, xerostomia, and retronasal smell intensity demonstrated significant inverse relationships with swallowing function. In addition, oral moisture levels demonstrated significant positive relationships with swallowing function. At 3 months, mucositis ratings maintained a significant, inverse relationship with swallow function. Taste and both orthonasal and retronasal smell intensity ratings demonstrated inverse relationships with measures of swallow function. Swallow functions and oral morbidities deteriorate significantly following CRT with incomplete recovery at 3 months post treatment. Furthermore, different patterns of relationships between swallow function measures and oral morbidities were obtained at the 6-week versus the 3-month assessment point suggesting that different mechanisms may contribute to the development versus the maintenance of dysphagia over the trajectory of treatment in these patients.


Asunto(s)
Quimioradioterapia/efectos adversos , Trastornos de Deglución/etiología , Neoplasias de Cabeza y Cuello/terapia , Enfermedades de la Boca/etiología , Anciano , Deglución/efectos de los fármacos , Deglución/efectos de la radiación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mucositis/etiología , Estudios Prospectivos , Factores de Tiempo , Resultado del Tratamiento , Xerostomía/etiología
9.
J Neurosci ; 36(42): 10769-10781, 2016 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-27798132

RESUMEN

Designer receptors exclusively activated by designer drugs (DREADDs) are an advanced experimental tool that could potentially provide a novel approach to pain management. In particular, expression of an inhibitory (Gi-coupled) DREADD in nociceptors might enable ligand-dependent analgesia. To test this possibility, TRPV1-cre mice were used to restrict expression of Gi-DREADDs to predominantly C-fibers. Whereas baseline heat thresholds in both male and female mice expressing Gi-DREADD were normal, 1 mg/kg clozapine-N-oxide (CNO) produced a significant 3 h increase in heat threshold that returned to baseline by 5 h after injection. Consistent with these behavioral results, CNO decreased action potential firing in isolated sensory neurons from Gi-DREADD mice. Unexpectedly, however, the expression of Gi-DREADD in sensory neurons caused significant changes in voltage-gated Ca2+ and Na+ currents in the absence of CNO, as well as an increase in Na+ channel (NaV1.7) expression. Furthermore, CNO-independent excitatory and inhibitory second-messenger signaling was also altered in these mice, which was associated with a decrease in the analgesic effect of endogenous inhibitory G-protein-coupled receptor activation. These results highlight the potential of this exciting technology, but also its limitations, and that it is essential to identify the underlying mechanisms for any observed behavioral phenotypes. SIGNIFICANCE STATEMENT: DREADD technology is a powerful tool enabling manipulation of activity and/or transmitter release from targeted cell populations. The purpose of this study was to determine whether inhibitory DREADDs in nociceptive afferents could be used to produce analgesia, and if so, how. DREADD activation produced a ligand-dependent analgesia to heat in vivo and a decrease in neuronal firing at the single-cell level. However, we observed that expression of Gi-DREADD also causes ligand-independent changes in ion channel activity and second-messenger signaling. These findings highlight both the potential and the limitations of this exciting technology as well as the necessity to identify the mechanisms underlying any observed phenotype.


Asunto(s)
Analgesia , Drogas de Diseño/farmacología , Nervios Periféricos/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Canales de Calcio/efectos de los fármacos , Clozapina/farmacología , Femenino , Masculino , Ratones , Nociceptores/efectos de los fármacos , Dimensión del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Sistemas de Mensajero Secundario/efectos de los fármacos , Canales Catiónicos TRPV/efectos de los fármacos
10.
Dev Biol ; 414(2): 149-60, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27151208

RESUMEN

Auditory information is initially processed in the cochlear nuclei before being relayed to the brain. The cochlear nuclei are subdivided into dorsal, anterior ventral, and posterior ventral domains, each containing several subtypes of neurons that are thought to play discrete roles in the processing of sound. However, the ontogeny of these neurons is poorly understood, and this gap in knowledge hampers efforts to understand the basic neural circuitry of this nucleus. Here, we reveal that Bhlhb5 is expressed in both excitatory (unipolar brush cells) and inhibitory neurons (cartwheel cells) of the DCN during development. To gain genetic access to Bhlhb5-expressing neurons in the DCN, we generated a Bhlhb5::flpo knockin allele. Using an intersectional genetic strategy, we labeled cartwheel cells, thereby providing proof of concept that subpopulations of Bhlhb5-expressing neurons can be genetically targeted. Moreover, fate-mapping experiments using this allele revealed that Bhlhb5 is required for the proper development of the DCN, since mice lacking Bhlhb5 showed a dramatically diminished number of neurons, including unipolar brush and cartwheel cells. Intriguingly, the Bhlhb5::flpo allele also genetically labels numerous other regions of the nervous system that process sensory input, including the dorsal horn, the retina, and the nucleus of the lateral olfactory tract, hinting at a more general role for Bhlhb5 in the development of neurons that mediate sensory integration.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Núcleo Coclear/crecimiento & desarrollo , Células Receptoras Sensoriales/metabolismo , Alelos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Recuento de Células , Linaje de la Célula , Núcleo Coclear/embriología , Núcleo Coclear/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Proteínas Luminiscentes/análisis , Ratones , Ratones Noqueados , Bulbo Olfatorio/metabolismo , Factor de Transcripción PAX6/metabolismo , Células del Asta Posterior/metabolismo , Retina/metabolismo
11.
Genesis ; 54(11): 593-601, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27712014

RESUMEN

The Neurokinin 1 Receptor (NK1R), which binds Substance P, is expressed in discrete populations of neurons throughout the nervous system, where it has numerous roles including the modulation of pain and affective behaviors. Here, we report the generation of a NK1R-CreER knockin allele, in which CreERT2 replaces the coding sequence of the TACR1 gene (encoding NK1R) in order to gain genetic access to these cells. We find that the NK1R-CreER allele mediates recombination in many regions of the nervous system that are important in pain and anxiety including the amygdala, hypothalamus, frontal cortex, raphe nucleus, and dorsal horn of the spinal cord. Other cell types that are labeled by this allele include amacrine cells in the retina and fibroblasts in the skin. Thus, the NK1R-CreER mouse line is a valuable new tool for conditional gene manipulation enabling the visualization and manipulation of cells that express NK1R.


Asunto(s)
Neuronas/metabolismo , Dolor/genética , Receptores de Neuroquinina-1/genética , Animales , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Integrasas/genética , Ratones , Dolor/patología , Receptores de Neuroquinina-1/metabolismo , Transducción de Señal/genética , Médula Espinal/crecimiento & desarrollo , Médula Espinal/metabolismo , Sustancia P/genética , Sustancia P/metabolismo
12.
Genesis ; 54(1): 29-37, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26575788

RESUMEN

The kappa opioid receptor (KOR) has numerous important roles in the nervous system including the modulation of mood, reward, pain, and itch. In addition, KOR is expressed in many non-neuronal tissues. However, the specific cell types that express KOR are poorly characterized. Here, we report the development of a KOR-Cre knockin allele, which provides genetic access to cells that express KOR. In this mouse, Cre recombinase (Cre) replaces the initial coding sequence of the Opkr1 gene (encoding the kappa opioid receptor). We demonstrate that the KOR-Cre allele mediates recombination by embryonic day 14.5 (E14.5). Within the brain, KOR-Cre shows expression in numerous areas including the cerebral cortex, nucleus accumbens and striatum. In addition, this allele is expressed in epithelium and throughout many regions of the body including the heart, lung, and liver. Finally, we reveal that KOR-Cre mediates recombination of a subset of bipolar and amacrine cells in the retina. Thus, the KOR-Cre mouse line is a valuable new tool for conditional gene manipulation to enable the study of KOR.


Asunto(s)
Técnicas de Transferencia de Gen , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Animales , Encéfalo/metabolismo , Femenino , Integrasas/química , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Receptores Opioides kappa/biosíntesis , Transducción de Señal
14.
Handb Exp Pharmacol ; 226: 191-206, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25861781

RESUMEN

Recent studies have made significant progress in the knowledge of how itch sensation is processed, especially the molecular identity of neurons involved in itch signaling, both in the dorsal root ganglion and spinal cord. Despite these advances, the organization of these neurons in dorsal spinal cord circuits and how they interact with other somatosensory modalities, such as pain or temperature, remain relatively unexplored. Recent work from our lab and others has begun to shed light on these questions and will be the focus of this chapter. Here we describe the discovery of B5-I neurons, a population of inhibitory interneurons that function to inhibit itch, and review the evidence that these neurons mediate the inhibition of itch by counter stimuli. These studies are helping to solve the long-standing question of why itch makes us scratch.


Asunto(s)
Interneuronas/fisiología , Prurito/fisiopatología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Humanos , Ratones , Prurito/prevención & control , Receptores Opioides kappa/agonistas , Sensación
15.
Gerontol Geriatr Med ; 10: 23337214241234737, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38410616

RESUMEN

Objective: This study tested the hypothesis that healthy aging attenuates cognitive practice effects and, consequently, limits the familiarity-associated reductions in heart rate (HR) and breathing frequency (BF) responses during retesting. Methods: Twenty-one cognitively normal older and younger adults (65 ± 2 vs. 26 ± 1 years old) participated in the study. Mini-Mental State Examination (MMSE), Digit-Span-Test (DST), Trail Making Test (TMT-B), and California Verbal Learning Test (CVLT-II) were administered twice at 3-week intervals, while HR and BF were monitored by electrocardiography and plethysmography, respectively. Results: Cognitive performances were not affected by the age factor, and the retest factor only affected CVLT-II. HR and BF increased only in the younger adults (p < .01) during cognitive tests; retesting attenuated these responses (retest factor p < .01). Long-delay free-recall in CVLT-II was unchanged in cognitively normal older versus younger adults. Healthy aging did not diminish short-term memory assessed by DST and CVLT-II short-delay or long-delay free-recalls. Conclusions: Only CVLT-II, but not MMSE, DST or TMT-B, demonstrated cognitive retesting practice effects in the younger and older adults. Cognitive testing at 3-week intervals in cognitively normal older and younger subjects revealed divergent cardiorespiratory responses to MMSE, DST, and TMT-B cognitive testing, particularly HR, which increased only in younger adults and to a lesser extent during retesting despite the absence of practice effects.

16.
Cell Rep ; 43(3): 113829, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38421871

RESUMEN

The nature of spinal output pathways that convey nociceptive information to the brain has been the subject of controversy. Here, we provide anatomical, molecular, and functional characterizations of two distinct anterolateral pathways: one, ascending in the lateral spinal cord, triggers nociceptive behaviors, and the other one, ascending in the ventral spinal cord, when inhibited, leads to sensorimotor deficits. Moreover, the lateral pathway consists of at least two subtypes. The first is a contralateral pathway that extends to the periaqueductal gray (PAG) and thalamus; the second is a bilateral pathway that projects to the bilateral parabrachial nucleus (PBN). Finally, we present evidence showing that activation of the contralateral pathway is sufficient for defensive behaviors such as running and freezing, whereas the bilateral pathway is sufficient for attending behaviors such as licking and guarding. This work offers insight into the complex organizational logic of the anterolateral system in the mouse.


Asunto(s)
Núcleos Parabraquiales , Médula Espinal , Ratones , Animales , Médula Espinal/fisiología , Tálamo/fisiología , Sustancia Gris Periacueductal/fisiología , Vías Nerviosas/fisiología
17.
J Am Geriatr Soc ; 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38828999

RESUMEN

BACKGROUND: Healthcare and community collaborations have the potential to address health-related social needs. We examined the implementation of an educational initiative and collaborative intervention between a geriatric clinic and Area Agency on Aging (AAA) to enhance age-friendly care for a Hispanic patient population. METHODS: As part of a Health Resources and Services Administration (HRSA)-funded Geriatric Workforce Enhancement Program, a geriatric clinic partnered with AAA to embed an English- and Spanish-speaking Social Service Coordinator (SSC). The SSC met with patients during new and annual visits or by referral to address What Matters and Mentation in the patient's primary language, provide education, and make social service referrals. Patients aged 60 and older, who received SSC services during a 12-month period, were defined as the intervention group (n = 112). Using a retrospective chart review, we compared them to a non-intervention group (n = 228) that received primary care. We examined available demographic and clinical data within the age-friendly areas of What Matters and Mentation. Measures included cognitive health screenings, advance care planning, patient education, and community referrals. RESULTS: Most of the intervention groups were eligible for AAA services and had the opportunity for service referrals to address identified needs. A higher proportion of patients within the intervention group completed screenings for cognitive health and advance care planning discussions. CONCLUSION: Interagency partnerships between ambulatory care settings and community-based organizations have the potential to expand access to linguistically and culturally competent age-friendly primary care for older adults.

18.
Cell Rep ; 43(4): 113970, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38512868

RESUMEN

To meet the high energy demands of brain function, cerebral blood flow (CBF) parallels changes in neuronal activity by a mechanism known as neurovascular coupling (NVC). However, which neurons play a role in mediating NVC is not well understood. Here, we identify in mice and humans a specific population of cortical GABAergic neurons that co-express neuronal nitric oxide synthase and tachykinin receptor 1 (Tacr1). Through whole-tissue clearing, we demonstrate that Tacr1 neurons extend local and long-range projections across functionally connected cortical areas. We show that whisker stimulation elicited Tacr1 neuron activity in the barrel cortex through feedforward excitatory pathways. Additionally, through optogenetic experiments, we demonstrate that Tacr1 neurons are instrumental in mediating CBF through the relaxation of mural cells in a similar fashion to whisker stimulation. Finally, by electron microscopy, we observe that Tacr1 processes contact astrocytic endfeet. These findings suggest that Tacr1 neurons integrate cortical activity to mediate NVC.


Asunto(s)
Acoplamiento Neurovascular , Animales , Ratones , Acoplamiento Neurovascular/fisiología , Humanos , Neuronas/metabolismo , Neuronas/fisiología , Vibrisas/fisiología , Ratones Endogámicos C57BL , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Masculino , Corteza Cerebral/fisiología , Corteza Cerebral/irrigación sanguínea , Circulación Cerebrovascular/fisiología , Óxido Nítrico Sintasa de Tipo I/metabolismo
19.
Dermatol Ther (Heidelb) ; 14(6): 1615-1631, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38814433

RESUMEN

INTRODUCTION: Skin involvement in patients with psoriatic arthritis (PsA) worsens the severity and burden of disease. Ixekizumab (IXE), a selective interleukin (IL)-17A antagonist, was compared to placebo (PBO) in the SPIRIT-P1 (NCT01695239) and SPIRIT-P2 (NCT02349295) studies in patients with PsA and evidence of plaque psoriasis. This post hoc analysis reports musculoskeletal, skin, and nail outcomes through week 24 in patients from SPIRIT-P1 and SPIRIT-P2, stratified by mild, moderate, or psoriasis at baseline. METHODS: This post hoc analysis pooled patients from SPIRIT-P1 and SPIRIT-P2 who were randomly assigned to PBO or IXE 80 mg every 4 weeks (Q4W) or every 2 weeks (Q2W). Efficacy outcomes were analyzed through week 24 by baseline psoriasis severity, defined by percent body surface area (BSA) affected; mild = BSA < 3%, moderate = 3% ≤ BSA ≤ 10%, severe = BSA > 10%. The primary outcomes assessed were the proportion of patients achieving American College of Rheumatology (ACR)20, ACR50, and ACR70 responses. Secondary outcomes included musculoskeletal, disease activity, skin and nail, and health-related quality-of-life measures. RESULTS: Similar proportions of patients achieved ACR20/ACR50/ACR70 over time across all severity subgroups and treatment arms. More than one-third of IXE-treated patients achieved ACR20 at week 4, or ACR50 at week 24, with no significant differences according to psoriasis severity at baseline. Disease activity outcomes were similar through week 24 with both IXEQ4W and IXEQ2W, regardless of psoriasis severity at baseline. There were no significant differences over 24 weeks in the proportions of IXE-treated patients with mild, moderate, or severe baseline psoriasis who achieved Minimal Disease Activity (MDA). Across all severity subgroups, IXE demonstrated Psoriasis Area Severity Index 100 response as early as week 4, and approximately one-third of IXE-treated patients achieved total skin clearance at week 24. CONCLUSION: IXE demonstrated rapid and consistent efficacy in joint, skin, and nail for patients with PsA, regardless of baseline psoriasis severity. TRIAL REGISTRATION: SPIRIT-P1 (NCT01695239), SPIRIT-P2 (NCT02349295).

20.
J Pain ; 24(2): 264-272, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36464136

RESUMEN

Opioid signaling has been shown to be critically important in the neuromodulation of sensory circuits in the superficial spinal cord. Agonists of the mu-opioid receptor (MOR) elicit itch, whereas agonists of the kappa-opioid receptor (KOR) have been shown to inhibit itch. Despite the clear roles of MOR and KOR for the modulation itch, whether the delta-opioid receptor (DOR) is involved in the regulation of itch remained unknown. Here, we show that intrathecal administration of DOR agonists suppresses chemical itch and that intrathecal application of DOR antagonists is sufficient to evoke itch. We identify that spinal enkephalin neurons co-express neuropeptide Y (NPY), a peptide previously implicated in the inhibition of itch. In the spinal cord, DOR overlapped with both the NPY receptor (NPY1R) and KOR, suggesting that DOR neurons represent a site for convergent itch information in the dorsal horn. Lastly, we found that neurons co-expressing DOR and KOR showed significant Fos induction following pruritogen-evoked itch. These results uncover a role for DOR in the modulation of itch in the superficial dorsal horn. PERSPECTIVE: This article reveals the role of the delta-opioid receptor in itch. Intrathecal administration of delta agonists suppresses itch whereas the administration of delta antagonists is sufficient to induce itch. These studies highlight the importance of delta-opioid signaling for the modulation of itch behaviors, which may represent new targets for the management of itch disorders.


Asunto(s)
Analgésicos Opioides , Receptores Opioides delta , Ratas , Animales , Analgésicos Opioides/farmacología , Ratas Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides kappa/agonistas , Asta Dorsal de la Médula Espinal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA