Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros










Intervalo de año de publicación
1.
Glia ; 72(6): 1054-1066, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38450799

RESUMEN

Neurons in sensory ganglia are wrapped completely by satellite glial cells (SGCs). One putative function of SGCs is to regulate the neuronal microenvironment, but this role has received only little attention. In this study we investigated whether the SGC envelope serves a barrier function and how SGCs may control the neuronal microenvironment. We studied this question on short-term (<24 h) cell cultures of dorsal root ganglia and trigeminal ganglia from adult mice, which contain neurons surrounded with SGCs, and neurons that are not. Using calcium imaging, we measured neuronal responses to molecules with established actions on sensory neurons. We found that neurons surrounded by SGCs had a smaller response to molecules such as adenosine triphosphate (ATP), glutamate, GABA, and bradykinin than neurons without glial cover. When we inhibited the activity of NTPDases, which hydrolyze the ATP, and also when we inhibited the glutamate and GABA transporters on SGCs, this difference in the neuronal response was no longer observed. We conclude that the SGC envelope does not hinder diffusional passage, but acts as a metabolic barrier that regulates the neuronal microenvironment, and can protect the neurons and modulate their activity.


Asunto(s)
Neuroglía , Neuronas , Animales , Ratones , Neuroglía/metabolismo , Ganglios Sensoriales , Ganglios Espinales , Glutamatos/metabolismo , Adenosina Trifosfato/metabolismo , Células Satélites Perineuronales/metabolismo
2.
Brain Res ; 1829: 148809, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38354998

RESUMEN

The sympathetic ganglia represent a final motor pathway that mediates homeostatic "fight and flight" responses in the visceral organs. Satellite glial cells (SGCs) form a thin envelope close to the neuronal cell body and synapses in the sympathetic ganglia. This unique morphological feature suggests that neurons and SGCs form functional units for regulation of sympathetic output. In the present study, we addressed whether SGC-specific markers undergo age-dependent changes in the postnatal development of rat sympathetic ganglia. We found that fatty acid-binding protein 7 (FABP7) is an early SGC marker, whereas the S100B calcium-binding protein, inwardly rectifying potassium channel, Kir4.1 and small conductance calcium-activated potassium channel, SK3 are late SGC markers in the postnatal development of sympathetic ganglia. Unlike in sensory ganglia, FABP7 + SGC was barely detectable in adult sympathetic ganglia. The expression of connexin 43, a gap junction channel gradually increased with age, although it was detected in both SGCs and neurons in sympathetic ganglia. Glutamine synthetase was expressed in sensory, but not sympathetic SGCs. Unexpectedly, the sympathetic SGCs expressed a water-selective channel, aquaporin 1 instead of aquaporin 4, a pan-glial marker. However, aquaporin 1 was not detected in the SGCs encircling large neurons. Nerve injury and inflammation induced the upregulation of glial fibrillary acidic protein, suggesting that this protein is a hall marker of glial activation in the sympathetic ganglia. In conclusion, our findings provide basic information on the in vivo profiles of specific markers for identifying sympathetic SGCs at different stages of postnatal development in both healthy and diseased states.


Asunto(s)
Neuroglía , Células Satélites Perineuronales , Ratas , Animales , Células Satélites Perineuronales/metabolismo , Neuroglía/metabolismo , Ganglios Simpáticos , Neuronas , Proteína de Unión a los Ácidos Grasos 7/metabolismo , Ganglios Espinales/metabolismo
3.
Int J Mol Sci ; 24(21)2023 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-37958541

RESUMEN

Satellite glial cells (SGCs), enveloping primary sensory neurons' somas in the dorsal root ganglion (DRG), contribute to neuropathic pain upon nerve injury. Glial fibrillary acidic protein (GFAP) serves as an SGC activation marker, though its DRG satellite cell specificity is debated. We employed the hGFAP-CFP transgenic mouse line, designed for astrocyte studies, to explore its expression within the peripheral nervous system (PNS) after spared nerve injury (SNI). We used diverse immunostaining techniques, Western blot analysis, and electrophysiology to evaluate GFAP+ cell changes. Post-SNI, GFAP+ cell numbers increased without proliferation, and were found near injured ATF3+ neurons. GFAP+ FABP7+ SGCs increased, yet 75.5% of DRG GFAP+ cells lacked FABP7 expression. This suggests a significant subset of GFAP+ cells are non-myelinating Schwann cells (nmSC), indicated by their presence in the dorsal root but not in the ventral root which lacks unmyelinated fibres. Additionally, patch clamp recordings from GFAP+ FABP7-cells lacked SGC-specific Kir4.1 currents, instead displaying outward Kv currents expressing Kv1.1 and Kv1.6 channels specific to nmSCs. In conclusion, this study demonstrates increased GFAP expression in two DRG glial cell subpopulations post-SNI: GFAP+ FABP7+ SGCs and GFAP+ FABP7- nmSCs, shedding light on GFAP's specificity as an SGC marker after SNI.


Asunto(s)
Neuralgia , Traumatismos del Sistema Nervioso , Animales , Ratones , Ganglios Espinales/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Neuroglía/metabolismo , Células Satélites Perineuronales/metabolismo , Neuralgia/metabolismo , Traumatismos del Sistema Nervioso/metabolismo
4.
Stem Cell Rev Rep ; 19(2): 358-367, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36242721

RESUMEN

Satellite glial cells (SGCs) that surround sensory neurons in the peripheral nervous system ganglia originate from neural crest cells. Although several studies have focused on SGCs, the origin and characteristics of SGCs are unknown, and their lineage remains unidentified. Traditionally, it has been considered that SGCs regulate the environment around neurons under pathological conditions, and perform functions of supporting, nourishing, and protecting neurons. However, recent studies demonstrated that SGCs may have the characteristics of stem cells. After nerve injury, SGCs up-regulate the expression of stem cell markers and can differentiate into functional sensory neurons. Moreover, SGCs express several markers of Schwann cell precursors and Schwann cells, such as CDH19, MPZ, PLP1, SOX10, ERBB3, and FABP7. Schwann cell precursors have also been proposed as a potential source of neurons in the peripheral nervous system. The similarity in function and markers suggests that SGCs may represent a subgroup of Schwann cell precursors. Herein, we discuss the roles and functions of SGCs, and the lineage relationship between SGCs and Schwann cell precursors. We also describe a new perspective on the roles and functions of SGCs. In the DRG located on the posterior root of spinal nerves, satellite glial cells wrap around each sensory neuron to form an anatomically and functionally distinct unit with the sensory neurons. Following nerve injury, satellite glial cells up-regulate the expression of progenitor markers, and can differentiate into neurons.


Asunto(s)
Neuroglía , Células Satélites Perineuronales , Células Satélites Perineuronales/metabolismo , Neuroglía/metabolismo , Células de Schwann , Células Receptoras Sensoriales
5.
Cells ; 11(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35159321

RESUMEN

Injury or inflammation in the peripheral branches of neurons of sensory ganglia causes changes in neuronal properties, including excessive firing, which may underlie chronic pain. The main types of glial cell in these ganglia are satellite glial cells (SGCs), which completely surround neuronal somata. SGCs undergo activation following peripheral lesions, which can enhance neuronal firing. How neuronal injury induces SGC activation has been an open question. Moreover, the mechanisms by which the injury is signaled from the periphery to the ganglia are obscure and may include electrical conduction, axonal and humoral transport, and transmission at the spinal level. We found that peripheral inflammation induced SGC activation and that the messenger between injured neurons and SGCs was nitric oxide (NO), acting by elevating cyclic guanosine monophosphate (cGMP) in SGCs. These results, together with work from other laboratories, indicate that a plausible (but not exclusive) mechanism for neuron-SGCs interactions can be formulated as follows: Firing due to peripheral injury induces NO formation in neuronal somata, which diffuses to SGCs. This stimulates cGMP synthesis in SGCs, leading to their activation and to other changes, which contribute to neuronal hyperexcitability and pain. Other mediators such as proinflammatory cytokines probably also contribute to neuron-SGC communications.


Asunto(s)
Dolor Crónico , Células Satélites Perineuronales , Dolor Crónico/metabolismo , Ganglios Sensoriales , Humanos , Inflamación/metabolismo , Neuroglía/metabolismo , Células Satélites Perineuronales/metabolismo
6.
Brain Res ; 1760: 147384, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33631206

RESUMEN

Neurons and satellite glial cells (SGCs) in sensory ganglia maintain bidirectional communications that are believed to be largely mediated by chemical messengers. Nerve injury leads to SGC activation, which was proposed to be mediated by nitric oxide (NO) released from active neurons, but evidence for this is lacking. Here we tested the idea that increased neuronal firing is a major factor in NO release. We activated neurons in isolated dorsal root and trigeminal ganglia from mice with capsaicin (5 µM), which acts on transient receptor potential vanilloid type 1 (TRPV1) channels in small neurons. We found that capsaicin induced SGC activation, as assayed by glial fibrillary acidic protein (GFAP) upregulation, and an NO-donor had a similar effect. Incubating the ganglia in capsaicin in the presence of the NO-synthase inhibitor L-NAME (100 µM) prevented the GFAP upregulation. We also found that capsaicin caused an increase in SGC-SGC coupling, which was shown previously to accompany SGC activation. To test the contribution of ATP to the actions of capsaicin, we incubated the ganglia with capsaicin in the presence of P2 purinergic receptor inhibitor suramin (100 µM), which prevented the capsaicin-induced GFAP upregulation. Size analysis indicated that although capsaicin acts mainly on small neurons, SGCs around neurons of all sizes were affected by capsaicin, suggesting a spread of signals from small neurons to neighboring cells. We conclude that neuronal excitation leads to NO release, which induces SGCs activation. It appears that ATP participates in NO's action, possibly by interaction with TRPV1 channels.


Asunto(s)
Comunicación Celular/fisiología , Ganglios Espinales/metabolismo , Neuronas/metabolismo , Células Satélites Perineuronales/metabolismo , Ganglio del Trigémino/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Neurotransmisores/metabolismo , Óxido Nítrico/metabolismo
7.
J Neurochem ; 157(3): 781-801, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33345310

RESUMEN

Perineuronal nets (PNNs) are presumed to limit plasticity in adult animals. Ischaemic stroke results in the massive breakdown of PNNs resulting in rejuvenating states of neuronal plasticity, but the mechanisms of this phenomenon are largely unknown. As hyaluronic acid (HA) is the structural backbone of PNNs, we hypothesized that these changes are a consequence of the altered expression of HA metabolism enzymes. Additionally, we investigated whether early hyaluronidase inhibition interferes with post-stroke PNN reduction and behavioural recovery. We investigated the mRNA/protein expression of these enzymes in the perilesional, remote and contralateral cortical regions in mice at different time points after photothrombosis, using quantitative real-time polymerase chain reaction and immunofluorescence. A skilled reaching test was employed to test hyaluronidase inhibitor L-ascorbic acid 6-hexadecanoate influence on post-stroke recovery. We found the simultaneous up-regulation of mRNA of HA synthesizing and degrading enzymes in the perilesional area early after stroke, suggesting an acceleration of HA turnover in ischaemic animals. Immunostaining revealed differential cellular localization of enzymes, with hyaluronidase 1 in astrocytes and hyaluronan synthase 2 in astrocytes and neurons, and post-stroke up-regulation of both of them in astrocytes. ß-glucuronidase was observed in neurons but post-stroke up-regulation occurred in microglia. Inhibition of hyaluronidase activity early after stroke resulted in improved performance in skilled reaching test, without affecting the numbers of PNNs. These results suggest that after stroke, a substantial reorganization of polysaccharide content occurs, and interfering with this process at early time has a beneficial effect on recovery.


Asunto(s)
Encéfalo/patología , Inhibidores Enzimáticos/uso terapéutico , Hialuronoglucosaminidasa/antagonistas & inhibidores , Accidente Cerebrovascular Isquémico/patología , Accidente Cerebrovascular Isquémico/terapia , Animales , Astrocitos/metabolismo , Femenino , Glucuronidasa/metabolismo , Hialuronano Sintasas/metabolismo , Ácido Hialurónico/biosíntesis , Ácido Hialurónico/metabolismo , Accidente Cerebrovascular Isquémico/psicología , Ratones , Ratones Endogámicos C57BL , Destreza Motora , Neuronas/metabolismo , Cultivo Primario de Células , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Recuperación de la Función , Células Satélites Perineuronales/metabolismo , Trombosis
8.
Biomed Pharmacother ; 127: 110187, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32361638

RESUMEN

Increasing evidence suggests that activation of satellite glia cells (SGCs) in sensory ganglia play important roles in the development of neuropathic pain. The present study aimed to investigate the involvement of SGC activation in a novel model of motor nerve injury induced pain hypersensitivity. The neuropathic pain model was established by cervical 8 ventral root avulsion (C8VA). Glial fibrillary acidic protein (GFAP) was used as a marker of SGC activation. Unilateral C8VA resulted in mechanical allodynia, but not thermal hyperalgesia in bilateral paws. Expectedly, SGCs were robustly activated on as early as 1 day and persisted for at least 7 days in the ipsilateral and contralateral dorsal root ganglia (DRG) of C6, C7 and C8 after C8VA. Double immunofluorescence showed that almost all the activated SGCs enveloped neurofilament 200 (NF200) positive myelinated neurons in DRG. Local application of fluorocitrate (FC), a glial metabolism inhibitor, significantly decreased the number of activated SGCs and alleviated bilateral mechanical allodynia. These results suggest that SGC activation contributed to ipsilateral and mirror-image pain hypersensitivity after C8VA. Inhibition of SGC activation represented a promising therapeutic strategy for the management of neuropathic pain following brachial plexus root avulsion.


Asunto(s)
Hiperalgesia/fisiopatología , Neuronas Motoras/patología , Neuralgia/fisiopatología , Células Satélites Perineuronales/metabolismo , Animales , Citratos/farmacología , Modelos Animales de Enfermedad , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Hiperalgesia/etiología , Masculino , Proteínas de Neurofilamentos/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Mol Cell Neurosci ; 105: 103499, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32389805

RESUMEN

Platinum-based chemotherapeutics still play an important role in cancer therapy, however, severe side effects, such as painful neuropathy, occur frequently. The pathophysiologic mechanisms depend on the applied chemotherapeutic agent and are still controversial. In addition to neuronal damage, disturbance of glial cell activity may contribute to neurotoxicity. Here, we focused on the effect of oxaliplatin on satellite glial cell (SGC) function and on the activity of the dorsal root ganglion (DRG) neurons. SGCs were isolated as high-purity cultures and treated with 1 and 10 µM oxaliplatin for 2, 4 and 24 h. Subsequently, glial fibrillary acid protein (GFAP), reactive oxygen species (ROS), Connexin-43 (Cx-43), and inward rectifier potassium channel 4.1 (Kir4.1) expression was determined by immunocytochemical staining (ICC) and Western blot analyses. Immunochemical staining and Western blot analysis showed an increase in the immune reactivity (IR) and protein levels of ROS, GFAP, and Cx-43. Furthermore, reduction of the IR and protein levels and current density were demonstrated using patch-clamp measurements, of Kir4.1 channels after oxaliplatin exposure. Cytokine release in SGCs was measured using enzyme-linked immunosorbent assays (ELISA) after oxaliplatin exposure and indicated an increased release of IL-6 and TNFα, while IL-1ß was decreased. The direct influence of SGC-secreted factors in the supernatant after oxaliplatin treatment led to the hyperexcitability of cultured DRG neurons. In summary, oxaliplatin has a direct impact on the modulation and function of different SGC proteins. Furthermore, SGC-released factors influence the excitability of sensory neurons, qualifying SGCs as potential targets for the prevention and treatment of oxaliplatin-induced polyneuropathy.


Asunto(s)
Ganglios Espinales/efectos de los fármacos , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Oxaliplatino/farmacología , Animales , Antineoplásicos/farmacología , Conexina 43/metabolismo , Ganglios Espinales/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Neuroglía/metabolismo , Oxaliplatino/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Células Satélites Perineuronales/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo
10.
Glia ; 68(7): 1375-1395, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32045043

RESUMEN

Satellite glial cells (SGCs) are homeostatic cells enveloping the somata of peripheral sensory and autonomic neurons. A wide variety of neuronal stressors trigger activation of SGCs, contributing to, for example, neuropathic pain through modulation of neuronal activity. However, compared to neurons and other glial cells of the nervous system, SGCs have received modest scientific attention and very little is known about SGC biology, possibly due to the experimental challenges associated with studying them in vivo and in vitro. Utilizing a recently developed method to obtain SGC RNA from dorsal root ganglia (DRG), we took a systematic approach to characterize the SGC transcriptional fingerprint by using next-generation sequencing and, for the first time, obtain an overview of the SGC injury response. Our RNA sequencing data are easily accessible in supporting information in Excel format. They reveal that SGCs are enriched in genes related to the immune system and cell-to-cell communication. Analysis of SGC transcriptional changes in a nerve injury-paradigm reveal a differential response at 3 days versus 14 days postinjury, suggesting dynamic modulation of SGC function over time. Significant downregulation of several genes linked to cholesterol synthesis was observed at both time points. In contrast, regulation of gene clusters linked to the immune system (MHC protein complex and leukocyte migration) was mainly observed after 14 days. Finally, we demonstrate that, after nerve injury, macrophages are in closer physical proximity to both small and large DRG neurons, and that previously reported injury-induced proliferation of SGCs may, in fact, be proliferating macrophages.


Asunto(s)
Ganglios Espinales/citología , Neuroglía/citología , Traumatismos de los Nervios Periféricos/metabolismo , Células Satélites Perineuronales/metabolismo , Animales , Comunicación Celular/fisiología , Femenino , Masculino , Ratones Endogámicos C57BL , Neuralgia/metabolismo , Neuroglía/metabolismo , Neuronas/citología , ARN/metabolismo , Células Satélites Perineuronales/fisiología
11.
Neural Plast ; 2019: 8924634, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31827499

RESUMEN

The pathophysiological processes leading to epilepsy are poorly understood. Understanding the molecular and cellular mechanisms involved in the onset of epilepsy is crucial for drug development. Epileptogenicity is thought to be associated with changes in synaptic plasticity; however, whether extracellular matrix molecules-known regulators of synaptic plasticity-are altered during epileptogenesis is unknown. To test this, we used a pentylenetetrazole- (PTZ-) kindling model mouse to investigate changes to hippocampal parvalbumin- (PV-) positive neurons, extracellular matrix molecules, and perineuronal nets (PNNs) after the last kindled seizure. We found an increase in Wisteria floribunda agglutinin- (WFA-) and Cat-315-positive PNNs and a decrease in PV-positive neurons not surrounded by PNNs, in the hippocampus of PTZ-kindled mice compared to control mice. Furthermore, the expression of WFA- and Cat-315-positive molecules increased in the extracellular space of PTZ-kindled mice. In addition, consistent with previous studies, astrocytes were activated in PTZ-kindled mice. We propose that the increase in PNNs after kindling decreases neuroplasticity in the hippocampus and helps maintain the neural circuit for recurrent seizures. This study shows that possibility of changes in extracellular matrix molecules due to astrocyte activation is associated with epilepticus in PTZ-kindled mice.


Asunto(s)
Matriz Extracelular/metabolismo , Hipocampo/metabolismo , Excitación Neurológica/fisiología , Red Nerviosa/metabolismo , Pentilenotetrazol/toxicidad , Células Satélites Perineuronales/metabolismo , Animales , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/patología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Excitación Neurológica/efectos de los fármacos , Excitación Neurológica/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/efectos de los fármacos , Red Nerviosa/patología , Células Satélites Perineuronales/efectos de los fármacos , Células Satélites Perineuronales/patología
12.
Neural Plast ; 2019: 6804575, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31772567

RESUMEN

The perineuronal net (PNN) is a mesh-like proteoglycan structure on the neuronal surface which is involved in regulating plasticity. The PNN regulates plasticity via multiple pathways, one of which is direct regulation of synapses through the control of AMPA receptor mobility. Since neuronal pentraxin 2 (Nptx2) is a known regulator of AMPA receptor mobility and Nptx2 can be removed from the neuronal surface by PNN removal, we investigated whether Nptx2 has a function in the PNN. We found that Nptx2 binds to the glycosaminoglycans hyaluronan and chondroitin sulphate E in the PNN. Furthermore, in primary cortical neuron cultures, the addition of NPTX2 to the culture medium enhances PNN formation during PNN development. These findings suggest Nptx2 as a novel PNN binding protein with a role in the mechanism of PNN formation.


Asunto(s)
Proteína C-Reactiva/metabolismo , Red Nerviosa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células Satélites Perineuronales/metabolismo , Corteza Visual/metabolismo , Animales , Células Cultivadas , Femenino , Red Nerviosa/química , Red Nerviosa/citología , Plasticidad Neuronal/fisiología , Neuronas/química , Neuronas/metabolismo , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Células Satélites Perineuronales/química , Corteza Visual/química , Corteza Visual/citología
13.
Gen Physiol Biophys ; 38(5): 407-416, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31595882

RESUMEN

The P2X7 receptor (P2X7R) plays an important role in inflammatory and neuropathic pain. Our recent study indicated that activation of P2X7R in microglial cells of spinal cord contributes to the inflammatory pain induced by BmK I, the major active compound from Buthus martensi Karsch (BmK). In the present study, we further investigated whether P2X7R in satellite glial cells (SGCs) of dorsal root ganglion (DRG) is involved in the BmK I-induced pain in rats. The results found that the expression of P2X7R in SGCs was increased in the ipsilateral side of L4-L5 DRGs after intraplantar injection of BmK I. Moreover, the expression of an inflammatory cytokine IL-1ß was increased in DRG after BmK I injection. Systemic administration of an inhibitor of P2X7R (A-438079) significantly inhibited both spontaneous and evoked nociceptive behaviors induced by BmK I. These results suggest that the P2X7R in SGCs of DRG might contribute to pain induced by toxins that sensitize peripheral sensory nerves.


Asunto(s)
Ganglios Espinales/patología , Dolor/inducido químicamente , Dolor/patología , Receptores Purinérgicos P2X7/metabolismo , Células Satélites Perineuronales/metabolismo , Venenos de Escorpión , Animales , Dolor/metabolismo , Ratas
14.
Brain Res Bull ; 149: 216-221, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31051227

RESUMEN

The satellite glial cells (SGCs) of the dorsal root ganglia (DRG) expressed P2X4 receptor. In this study, we investigated the abnormal sympathetic activity after myocardial ischemia (MI) involving P2X4 receptor in the cervical DRG SGC. The results showed that MI injury upregulated the P2X4 receptor mRNA and protein in DRG, and the upregulated P2X4 receptor was co-localized with glial fibrillary acidic protein (GFAP) in DRG SGCs. P2X4 short hairpin RNA (shRNA) treatment decreased the expression of P2X4 receptor, counteracted the upregulation of GFAP and IL-1ß and inhibited P38MAPK phosphorylation in DRG of MI rats. These results indicate that application of P2X4 shRNA may reduce P2X4-mediated nociceptive signal via inhibiting DRG afferents to alleviate the abnormal sympathetic activity induced by MI.


Asunto(s)
Ganglios Espinales/metabolismo , Ganglios Espinales/fisiopatología , Isquemia Miocárdica/fisiopatología , Neuroglía/metabolismo , Receptores Purinérgicos P2X4/metabolismo , Animales , Diabetes Mellitus Experimental/metabolismo , Neuropatías Diabéticas/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Isquemia Miocárdica/metabolismo , Neuralgia/metabolismo , Antagonistas del Receptor Purinérgico P2X/farmacología , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Células Satélites Perineuronales/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
J Cell Physiol ; 234(11): 21199-21210, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31032956

RESUMEN

Satellite glial cells (SGCs) activation in the trigeminal ganglia (TG) is critical in various abnormal orofacial sensation in nerve injury and inflammatory conditions. SGCs express several subtypes of P2 purinergic receptors contributing to the initiation and maintenance of neuropathic pain. The P2Y14 receptor, a G-protein-coupled receptor activated by uridine diphosphate (UDP)-glucose and other UDP sugars, mediates various physiologic events such as immune, inflammation, and pain. However, the expression, distribution, and function of P2Y14 receptor in SGCs remains largely unexplored. Our study reported the expression and functional identification of P2Y14 receptor in SGCs. SGCs were isolated from TG of rat, and the P2Y14 receptor expression was examined using immunofluorescence technique. Cell proliferation and viability were examined via cell counting kit-8 experiment. Immunofluorescence demonstrated the presence of P2Y14 receptor in SGCs. Immunofluorescence and western blot showed that UDP-glucose treatment upregulated glial fibrillary acid protein, a common marker for glial activation. Extracellular UDP-glucose enhanced the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38, which were both abolished by the P2Y14 receptor inhibitor (PPTN). Furthermore, quantitative reverse transcription polymerase chain reaction and enzyme-linked immunosorbent assay demonstrated that extracellular UDP-glucose significantly enhanced interleukin-1ß (IL-1ß) and chemokine CCL2 (CCL2) release, which was abolished by PPTN and significantly decreased by inhibitors of MEK/ERK (U0126) and p38 (SB202190). Our findings directly proved the functional presence of P2Y14 receptor in SGCs. It was also verified that P2Y14 receptor activation was involved in activating SGCs, phosphorylating MAPKs, and promoting the secretion of IL-1ß and CCL2 via ERK and p38 pathway.


Asunto(s)
Quimiocina CCL2/metabolismo , Interleucina-1beta/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Células Satélites Perineuronales/metabolismo , Animales , Masculino , Ratas , Ratas Sprague-Dawley
16.
Brain Res Bull ; 149: 240-250, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31034945

RESUMEN

Orofacial inflammation could activate satellite glial cells (SGCs) in the trigeminal ganglion (TG) to produce interleukin 1ß (IL-1ß) which plays crucial roles in the development of inflammatory pain. Recent studies have shown that gamma-amino butyric acid-B (GABAB) receptor could modulate the expression of inflammatory cytokines in microglia and astrocytes in the spinal cord. The objective of this study was to investigate whether GABAB receptors in TG SGCs attenuate inflammatory facial pain via mediating IL-1ß following inflammation and its mechanisms. Complete Freund's adjuvant (CFA) was injected into the whisker pad of rats to induce inflammation in vivo. Lipopolysaccharide (LPS) was added to culture medium to activate SGCs in vitro. Behavioral measures showed that microinjection of baclofen (a selective GABAB receptor agonist) into the TG ameliorated the mechanical allodynia of CFA-treated rats. Interestingly, baclofen pretreatment inhibited SGC activation and IL-1ß production, however, preserved the decreased expression of GABAB receptors in SGCs activated by CFA in vivo and LPS in vitro. In addition, baclofen suppressed the increased expression of p-NF- κ B p65, p-I κ Bα, and p-p38 MAPK, while reversed the decreased production of I κ Bα, and further enhanced the increased expression of p-ERK(1/2) in LPS-treated SGCs in vitro. Finally, those effects of baclofen were abolished by saclofen (a specific GABAB receptor antagonist) co-administration. Altogether, these results demonstrated for the first time that activation of GABAB receptor might inhibit IL-1ß production by suppressing NF- κ B and p38 MAPK signaling pathway activation and restore GABAB receptor expression in SGCs to attenuate inflammatory facial pain.


Asunto(s)
Dolor Facial/metabolismo , Interleucina-1beta/metabolismo , Receptores de GABA-B/metabolismo , Animales , Baclofeno/farmacología , Citocinas/metabolismo , Dolor Facial/fisiopatología , Agonistas de Receptores GABA-B/farmacología , Hiperalgesia/metabolismo , Inflamación , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Microglía/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Neuroglía/metabolismo , Neuroglía/fisiología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Células Satélites Perineuronales/metabolismo , Transducción de Señal/fisiología , Ganglio del Trigémino/metabolismo , Ácido gamma-Aminobutírico/metabolismo
17.
Glia ; 67(7): 1296-1307, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30801760

RESUMEN

Abnormal neuronal activity in sensory ganglia contributes to chronic pain. There is evidence that signals can spread between cells in these ganglia, which may contribute to this activity. Satellite glial cells (SGCs) in sensory ganglia undergo activation following peripheral injury and participate in cellular communication via gap junctions and chemical signaling. Nitric oxide (NO) is released from neurons in dorsal root ganglia (DRG) and induces cyclic GMP (cGMP) production in SCGs, but its role in SGC activation and neuronal excitability has not been explored. It was previously reported that induction of intestinal inflammation with dinitrobenzoate sulfonate (DNBS) increased gap junctional communications among SGCs, which contributed to neuronal excitability and pain. Here we show that DNBS induced SGC activation in mouse DRG, as assayed by glial fibrillary acidic protein upregulation. DNBS also upregulated cGMP level in SGCs, consistent with NO production. In vitro studies on intact ganglia from DNBS-treated mice showed that blocking NO synthesis inhibited both SGCs activation and cGMP upregulation, indicating an ongoing NO production. Application of NO donor in vitro induced SGC activation, augmented gap junctional communications, and raised neuronal excitability, as assessed by electrical recordings. The cGMP analog 8-Br-cGMP mimicked these actions, confirming the role of the NO-cGMP pathway in intraganglionic communications. NO also augmented Ca2+ waves propagation in DRG cultures. It is proposed that NO synthesis in DRG neurons increases after peripheral inflammation and that NO induces SGC activation, which in turn contributes to neuronal hyperexcitability. Thus, NO plays a major role in neuron-SGC communication.


Asunto(s)
Comunicación Celular/fisiología , Ganglios Espinales/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Óxido Nítrico/biosíntesis , Células Satélites Perineuronales/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Dinitrofluorobenceno/análogos & derivados , Dinitrofluorobenceno/farmacología , Femenino , Ganglios Espinales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Células Satélites Perineuronales/efectos de los fármacos
18.
Glia ; 67(6): 1062-1075, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30648289

RESUMEN

Chronic pain is one of the most prevalent chronic diseases in the world. The plastic changes of sensory neurons in dorsal root ganglia (DRG) have been extensively studied as the underlying periphery mechanism. Recent studies revealed that satellite cells, the major glial cells in DRG, also played important roles in the development/modulation of chronic pain. Whether DRG satellite glial cells generate new neurons as their counterparts in enteric nerve ganglia and carotid body do under pathological conditions remains poorly investigated. Here, we report that chronic pain induces proliferation and upregulation of progenitor markers in the sex-determining region Y-box 2 (Sox2)- and platelet-derived growth factor receptor alpha (PDGFRα)-positive satellite glial cells. BrdU incorporation assay revealed the generation of IB4- and CGRP-positive neurons, but not NF200-positive neurons in DRG ipsilateral to injury. Genetic fate tracings showed that PDGFRα-positive cells did not generate neurons, whereas Sox2-positive cells produced both IB4- and CGRP-positive neurons. Interestingly, glial fibrillary acidic protein-positive cells, a subpopulation of Sox2-positive satellites, only gave birth to IB4-positive neurons. Local persistent delivery of tetrodotoxin to the sciatic nerve trunk significantly reduced the pain-induced neurogenesis. Furthermore, patch-clamp studies demonstrated that these glia-derived new neurons could fire action potentials and respond to capsaicin. Taken together, our data demonstrated a chronic pain-induced nociceptive neurogenesis in DRG from Sox2-positive satellite cells, indicating a possible contribution of DRG neurogenesis to the pathology of chronic pain.


Asunto(s)
Dolor Crónico/metabolismo , Ganglios Espinales/metabolismo , Neurogénesis/fisiología , Factores de Transcripción SOXB1/biosíntesis , Células Satélites Perineuronales/metabolismo , Animales , Dolor Crónico/patología , Ganglios Espinales/química , Ganglios Espinales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción SOXB1/análisis , Células Satélites Perineuronales/química , Células Satélites Perineuronales/patología
19.
Neurosci Lett ; 695: 46-52, 2019 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28647288

RESUMEN

Enhanced expression and function of gap junctions and pannexin (Panx) channels have been associated with both peripheral and central mechanisms of pain sensitization. At the level of the sensory ganglia, evidence includes augmented gap junction and pannexin1 expression in glial cells and neurons in inflammatory and neuropathic pain models and increased synchrony and enhanced cross-excitation among sensory neurons by gap junction-mediated coupling. In spinal cord and in suprapinal areas, evidence is largely limited to increased expression of relevant proteins, although in several rodent pain models, hypersensitivity is reduced by treatment with gap junction/Panx1 channel blocking compounds. Moreover, targeted modulation of Cx43 expression was shown to modulate pain thresholds, albeit in somewhat contradictory ways, and mice lacking Panx1 expression globally or in specific cell types show depressed hyperalgesia. We here review the evidence for involvement of gap junctions and Panx channels in a variety of animal pain studies and then discuss ways in which gap junctions and Panx channels may mediate their action in pain processing. This discussion focusses on spread of signals among satellite glial cells, in particular intercellular Ca2+ waves, which are propagated through both gap junction and Panx1-dependent routes and have been associated with the phenomenon of spreading depression and the malady of migraine headache with aura.


Asunto(s)
Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Dolor/metabolismo , Animales , Ganglios Sensoriales/metabolismo , Ganglios Sensoriales/patología , Hiperalgesia/metabolismo , Hiperalgesia/patología , Neuralgia/metabolismo , Neuralgia/patología , Neuroglía/metabolismo , Neuroglía/patología , Dolor/patología , Células Satélites Perineuronales/metabolismo , Células Satélites Perineuronales/patología , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/patología
20.
J Neurosci Res ; 97(4): 393-401, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30450738

RESUMEN

Some chronic pain conditions in the orofacial region are common, the mechanisms underlying which are unresolved. Satellite glial cells (SGCs) are the glial cells of the peripheral nervous system. In the sensory ganglia, each neuronal body is surrounded by SGCs forming distinct functional units. The unique structural organization enables SGCs to communicate with each other and with their enwrapped neurons via a variety of ways. There is a growing body of evidence that SGCs can influence the level of neuronal excitability and are involved in the development and/or maintenance of pain. The aim of this review was to summarize the latest advances made about the implication of SGCs in orofacial pain. It may offer new targets for the development of orofacial pain treatment.


Asunto(s)
Comunicación Celular/fisiología , Dolor Facial/metabolismo , Neuralgia/metabolismo , Neuroglía/fisiología , Células Satélites Perineuronales/metabolismo , Ganglios Sensoriales/metabolismo , Humanos , Neuroglía/metabolismo , Neuronas/fisiología , Ganglio del Trigémino/fisiología , Nervio Trigémino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...