Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Añadir filtros








Intervalo de año
1.
Neuroscience Bulletin ; (6): 1188-1202, 2021.
Artículo en Chino | WPRIM | ID: wpr-951958

RESUMEN

Programmed cell death protein 1 (PD-1) is an immune checkpoint modulator and a major target of immunotherapy as anti-PD-1 monoclonal antibodies have demonstrated remarkable efficacy in cancer treatment. Accumulating evidence suggests an important role of PD-1 in the central nervous system (CNS). PD-1 has been implicated in CNS disorders such as brain tumors, Alzheimer’s disease, ischemic stroke, spinal cord injury, multiple sclerosis, cognitive function, and pain. PD-1 signaling suppresses the CNS immune response via resident microglia and infiltrating peripheral immune cells. Notably, PD-1 is also widely expressed in neurons and suppresses neuronal activity via downstream Src homology 2 domain-containing protein tyrosine phosphatase 1 and modulation of ion channel function. An improved understanding of PD-1 signaling in the cross-talk between glial cells, neurons, and peripheral immune cells in the CNS will shed light on immunomodulation, neuromodulation, and novel strategies for treating brain diseases.

2.
Neuroscience Bulletin ; (6): 13-21, 2018.
Artículo en Inglés | WPRIM | ID: wpr-777084

RESUMEN

Mounting evidence supports an important role of chemokines, produced by spinal cord astrocytes, in promoting central sensitization and chronic pain. In particular, CCL2 (C-C motif chemokine ligand 2) has been shown to enhance N-methyl-D-aspartate (NMDA)-induced currents in spinal outer lamina II (IIo) neurons. However, the exact molecular, synaptic, and cellular mechanisms by which CCL2 modulates central sensitization are still unclear. We found that spinal injection of the CCR2 antagonist RS504393 attenuated CCL2- and inflammation-induced hyperalgesia. Single-cell RT-PCR revealed CCR2 expression in excitatory vesicular glutamate transporter subtype 2-positive (VGLUT2) neurons. CCL2 increased NMDA-induced currents in CCR2/VGLUT2 neurons in lamina IIo; it also enhanced the synaptic NMDA currents evoked by dorsal root stimulation; and furthermore, it increased the total and synaptic NMDA currents in somatostatin-expressing excitatory neurons. Finally, intrathecal RS504393 reversed the long-term potentiation evoked in the spinal cord by C-fiber stimulation. Our findings suggest that CCL2 directly modulates synaptic plasticity in CCR2-expressing excitatory neurons in spinal lamina IIo, and this underlies the generation of central sensitization in pathological pain.


Asunto(s)
Animales , Femenino , Masculino , Ratones , Benzoxazinas , Farmacología , Usos Terapéuticos , Quimiocina CCL2 , Genética , Metabolismo , Farmacología , Fármacos actuantes sobre Aminoácidos Excitadores , Farmacología , Agonistas de Aminoácidos Excitadores , Farmacología , Adyuvante de Freund , Toxicidad , Hiperalgesia , Metabolismo , Potenciación a Largo Plazo , Fisiología , Proteínas Luminiscentes , Genética , Metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Mielitis , Quimioterapia , Metabolismo , Neuronas , Manejo del Dolor , Somatostatina , Genética , Metabolismo , Médula Espinal , Biología Celular , Compuestos de Espiro , Farmacología , Usos Terapéuticos , Proteína 2 de Transporte Vesicular de Glutamato , Genética , Metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores , Genética , Metabolismo
3.
Neuroscience Bulletin ; (6): 42-53, 2018.
Artículo en Inglés | WPRIM | ID: wpr-777083

RESUMEN

Increasing evidence suggests that cytokines and chemokines play crucial roles in chronic itch. In the present study, we evaluated the roles of tumor necrosis factor-alpha (TNF-α) and its receptors TNF receptor subtype-1 (TNFR1) and TNFR2 in acute and chronic itch in mice. Compared to wild-type (WT) mice, TNFR1-knockout (TNFR1-KO) and TNFR1/R2 double-KO (DKO), but not TNFR2-KO mice, exhibited reduced acute itch induced by compound 48/80 and chloroquine (CQ). Application of the TNF-synthesis inhibitor thalidomide and the TNF-α antagonist etanercept dose-dependently suppressed acute itch. Intradermal injection of TNF-α was not sufficient to evoke scratching, but potentiated itch induced by compound 48/80, but not CQ. In addition, compound 48/80 induced TNF-α mRNA expression in the skin, while CQ induced its expression in the dorsal root ganglia (DRG) and spinal cord. Furthermore, chronic itch induced by dry skin was reduced by administration of thalidomide and etanercept and in TNFR1/R2 DKO mice. Dry skin induced TNF-α expression in the skin, DRG, and spinal cord and TNFR1 expression only in the spinal cord. Thus, our findings suggest that TNF-α/TNFR1 signaling is required for the full expression of acute and chronic itch via peripheral and central mechanisms, and targeting TNFR1 may be beneficial for chronic itch treatment.


Asunto(s)
Animales , Masculino , Ratones , Cloroquina , Toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Etanercept , Usos Terapéuticos , Ganglios Espinales , Metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Prurito , Quimioterapia , Metabolismo , Patología , ARN Mensajero , Metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral , Genética , Receptores Tipo II del Factor de Necrosis Tumoral , Genética , Transducción de Señal , Piel , Metabolismo , Médula Espinal , Metabolismo , Talidomida , Usos Terapéuticos , Factores de Tiempo , Factor de Necrosis Tumoral alfa , Genética , Metabolismo , p-Metoxi-N-metilfenetilamina , Toxicidad
4.
Neuroscience Bulletin ; (6): 4-12, 2018.
Artículo en Inglés | WPRIM | ID: wpr-777078

RESUMEN

Voltage-gated sodium channels (Navs) play an important role in human pain sensation. However, the expression and role of Nav subtypes in native human sensory neurons are unclear. To address this issue, we obtained human dorsal root ganglion (hDRG) tissues from healthy donors. PCR analysis of seven DRG-expressed Nav subtypes revealed that the hDRG has higher expression of Nav1.7 (~50% of total Nav expression) and lower expression of Nav1.8 (~12%), whereas the mouse DRG has higher expression of Nav1.8 (~45%) and lower expression of Nav1.7 (~18%). To mimic Nav regulation in chronic pain, we treated hDRG neurons in primary cultures with paclitaxel (0.1-1 μmol/L) for 24 h. Paclitaxel increased the Nav1.7 but not Nav1.8 expression and also increased the transient Na currents and action potential firing frequency in small-diameter (<50 μm) hDRG neurons. Thus, the hDRG provides a translational model in which to study "human pain in a dish" and test new pain therapeutics.


Asunto(s)
Animales , Femenino , Humanos , Masculino , Ratones , Potenciales de Acción , Antineoplásicos Fitogénicos , Farmacología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores , Ganglios Espinales , Biología Celular , Regulación de la Expresión Génica , Técnicas In Vitro , Genética , Metabolismo , Neuronas , Metabolismo , Paclitaxel , Farmacología , Técnicas de Placa-Clamp , Especificidad de la Especie
5.
Neuroscience Bulletin ; (6): 98-108, 2018.
Artículo en Inglés | WPRIM | ID: wpr-777072

RESUMEN

Increasing evidence suggests that spinal microglia regulate pathological pain in males. In this study, we investigated the effects of several microglial and astroglial modulators on inflammatory and neuropathic pain following intrathecal injection in male and female mice. These modulators were the microglial inhibitors minocycline and ZVEID (a caspase-6 inhibitor) and the astroglial inhibitors L-α-aminoadipate (L-AA, an astroglial toxin) and carbenoxolone (a connexin 43 inhibitor), as well as U0126 (an ERK kinase inhibitor) and D-JNKI-1 (a c-Jun N-terminal kinase inhibitor). We found that spinal administration of minocycline or ZVEID, or Caspase6 deletion, reduced formalin-induced inflammatory and nerve injury-induced neuropathic pain primarily in male mice. In contrast, intrathecal L-AA reduced neuropathic pain but not inflammatory pain in both sexes. Intrathecal U0126 and D-JNKI-1 reduced neuropathic pain in both sexes. Nerve injury caused spinal upregulation of the astroglial markers GFAP and Connexin 43 in both sexes. Collectively, our data confirmed male-dominant microglial signaling but also revealed sex-independent astroglial signaling in the spinal cord in inflammatory and neuropathic pain.


Asunto(s)
Animales , Femenino , Masculino , Ratones , Ácido 2-Aminoadípico , Toxicidad , Antiinflamatorios , Usos Terapéuticos , Astrocitos , Patología , Carbenoxolona , Farmacología , Caspasa 6 , Metabolismo , Conexina 43 , Metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos , Farmacología , Proteína Ácida Fibrilar de la Glía , Metabolismo , Ratones Transgénicos , Microglía , Patología , Minociclina , Usos Terapéuticos , Neuralgia , Quimioterapia , Patología , Dimensión del Dolor , Compuestos de Fenilurea , Farmacología , Caracteres Sexuales , Médula Espinal , Patología , Factores de Tiempo
6.
Neuroscience Bulletin ; (6): 22-41, 2018.
Artículo en Inglés | WPRIM | ID: wpr-777048

RESUMEN

The voltage-gated Na channel subtype Nav1.7 is important for pain and itch in rodents and humans. We previously showed that a Nav1.7-targeting monoclonal antibody (SVmab) reduces Na currents and pain and itch responses in mice. Here, we investigated whether recombinant SVmab (rSVmab) binds to and blocks Nav1.7 similar to SVmab. ELISA tests revealed that SVmab was capable of binding to Nav1.7-expressing HEK293 cells, mouse DRG neurons, human nerve tissue, and the voltage-sensor domain II of Nav1.7. In contrast, rSVmab showed no or weak binding to Nav1.7 in these tests. Patch-clamp recordings showed that SVmab, but not rSVmab, markedly inhibited Na currents in Nav1.7-expressing HEK293 cells. Notably, electrical field stimulation increased the blocking activity of SVmab and rSVmab in Nav1.7-expressing HEK293 cells. SVmab was more effective than rSVmab in inhibiting paclitaxel-induced mechanical allodynia. SVmab also bound to human DRG neurons and inhibited their Na currents. Finally, potential reasons for the differential efficacy of SVmab and rSVmab and future directions are discussed.


Asunto(s)
Animales , Femenino , Humanos , Masculino , Ratones , Anticuerpos Monoclonales , Usos Terapéuticos , Biotina , Metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Ganglios Espinales , Biología Celular , Células HEK293 , Hibridomas , Química , Hiperalgesia , Quimioterapia , Ratones Endogámicos C57BL , Metabolismo , Química , Alergia e Inmunología , Metabolismo , Neuralgia , Quimioterapia , Metabolismo , Unión Proteica , Proteínas Recombinantes , Usos Terapéuticos , Células Receptoras Sensoriales , Fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA