Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Front Pain Res (Lausanne) ; 5: 1411879, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38693962
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.
Neural Regen Res ; 19(5): 1056-1063, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-37862208

RESUMEN

Satellite glial cells are unique glial cells that surround the cell body of primary sensory neurons. An increasing body of evidence suggests that in the presence of inflammation and nerve damage, a significant number of satellite glial cells become activated, thus triggering a series of functional changes. This suggests that satellite glial cells are closely related to the occurrence of chronic pain. In this review, we first summarize the morphological structure, molecular markers, and physiological functions of satellite glial cells. Then, we clarify the multiple key roles of satellite glial cells in chronic pain, including gap junction hemichannel Cx43, membrane channel Pannexin1, K channel subunit 4.1, ATP, purinergic P2 receptors, and a series of additional factors and their receptors, including tumor necrosis factor, glutamate, endothelin, and bradykinin. Finally, we propose that future research should focus on the specific sorting of satellite glial cells, and identify genomic differences between physiological and pathological conditions. This review provides an important perspective for clarifying mechanisms underlying the peripheral regulation of chronic pain and will facilitate the formulation of new treatment plans for chronic pain.

4.
Front Pain Res (Lausanne) ; 4: 1204057, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37325677

RESUMEN

Reactive oxygen species (ROS) are generated in nociceptive pathways in response to inflammation and injury. ROS are accumulated within the sensory ganglia following peripheral inflammation, but the functional role of intraganlionic ROS in inflammatory pain is not clearly understood. The aims of this study were to investigate whether peripheral inflammation leads to prolonged ROS accumulation within the trigeminal ganglia (TG), whether intraganglionic ROS mediate pain hypersensitivity via activation of TRPA1, and whether TRPA1 expression is upregulated in TG during inflammatory conditions by ROS. We demonstrated that peripheral inflammation causes excess ROS production within TG during the period when inflammatory mechanical hyperalgesia is most prominent. Additionally, scavenging intraganglionic ROS attenuated inflammatory mechanical hyperalgesia and a pharmacological blockade of TRPA1 localized within TG also mitigated inflammatory mechanical hyperalgesia. Interestingly, exogenous administration of ROS into TG elicited mechanical hyperalgesia and spontaneous pain-like responses via TRPA1, and intraganglionic ROS induced TRPA1 upregulation in TG. These results collectively suggest that ROS accumulation in TG during peripheral inflammation contributes to pain and hyperalgesia in a TRPA1 dependent manner, and that ROS further exacerbate pathological pain responses by upregulating TRPA1 expression. Therefore, any conditions that exacerbate ROS accumulation within somatic sensory ganglia can aggravate pain responses and treatments reducing ganglionic ROS may help alleviate inflammatory pain.

5.
Stem Cell Reports ; 18(5): 1155-1165, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37084722

RESUMEN

Here we describe a novel neuro-mesodermal assembloid model that recapitulates aspects of peripheral nervous system (PNS) development such as neural crest cell (NCC) induction, migration, and sensory as well as sympathetic ganglion formation. The ganglia send projections to the mesodermal as well as neural compartment. Axons in the mesodermal part are associated with Schwann cells. In addition, peripheral ganglia and nerve fibers interact with a co-developing vascular plexus, forming a neurovascular niche. Finally, developing sensory ganglia show response to capsaicin indicating their functionality. The presented assembloid model could help to uncover mechanisms of human NCC induction, delamination, migration, and PNS development. Moreover, the model could be used for toxicity screenings or drug testing. The co-development of mesodermal and neuroectodermal tissues and a vascular plexus along with a PNS allows us to investigate the crosstalk between neuroectoderm and mesoderm and between peripheral neurons/neuroblasts and endothelial cells.


Asunto(s)
Células Endoteliales , Células-Madre Neurales , Humanos , Células de Schwann , Axones , Mesodermo , Cresta Neural/fisiología
6.
Cell Rep ; 38(5): 110328, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108545

RESUMEN

Satellite glia are the major glial type found in sympathetic and sensory ganglia in the peripheral nervous system, and specifically, contact neuronal cell bodies. Sympathetic and sensory neurons differ in morphological, molecular, and electrophysiological properties. However, the molecular diversity of the associated satellite glial cells remains unclear. Here, using single-cell RNA sequencing analysis, we identify five different populations of satellite glia from sympathetic and sensory ganglia. We define three shared populations of satellite glia enriched in immune-response genes, immediate-early genes, and ion channels/ECM-interactors, respectively. Sensory- and sympathetic-specific satellite glia are differentially enriched for modulators of lipid synthesis and metabolism. Sensory glia are also specifically enriched for genes involved in glutamate turnover. Furthermore, satellite glia and Schwann cells can be distinguished by unique transcriptional signatures. This study reveals the remarkable heterogeneity of satellite glia in the peripheral nervous system.


Asunto(s)
Ganglios Sensoriales/metabolismo , Ganglios Espinales/metabolismo , Neuroglía/metabolismo , Células de Schwann/metabolismo , Animales , Ganglios Simpáticos/metabolismo , Humanos , Ratones , Neuronas/metabolismo , Neuronas Aferentes , Sistema Nervioso Periférico/metabolismo
7.
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
8.
Handb Exp Pharmacol ; 275: 53-90, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34796381

RESUMEN

Taste information is encoded in the gustatory nervous system much as in other sensory systems, with notable exceptions. The concept of adequate stimulus is common to all sensory modalities, from somatosensory to auditory, visual, and so forth. That is, sensory cells normally respond only to one particular form of stimulation, the adequate stimulus, such as photons (photoreceptors in the visual system), odors (olfactory sensory neurons in the olfactory system), noxious heat (nociceptors in the somatosensory system), etc. Peripheral sensory receptors transduce the stimulus into membrane potential changes transmitted to the brain in the form of trains of action potentials. How information concerning different aspects of the stimulus such as quality, intensity, and duration are encoded in the trains of action potentials is hotly debated in the field of taste. At one extreme is the notion of labeled line/spatial coding - information for each different taste quality (sweet, salty, sour, etc.) is transmitted along a parallel but separate series of neurons (a "line") that project to focal clusters ("spaces") of neurons in the gustatory cortex. These clusters are distinct for each taste quality. Opposing this are concepts of population/combinatorial coding and temporal coding, where taste information is encrypted by groups of neurons (circuits) and patterns of impulses within these neuronal circuits. Key to population/combinatorial and temporal coding is that impulse activity in an individual neuron does not provide unambiguous information about the taste stimulus. Only populations of neurons and their impulse firing pattern yield that information.


Asunto(s)
Células Receptoras Sensoriales , Gusto , Potenciales de Acción/fisiología , Humanos , Percepción , Gusto/fisiología
10.
Dev Dyn ; 250(10): 1450-1462, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33715274

RESUMEN

BACKGROUND: Eya2 expression during mouse development has been studied by in situ hybridization and it has been shown to be involved skeletal muscle development and limb formation. Here, we generated Eya2 knockout (Eya2- ) and a lacZ knockin reporter (Eya2lacZ ) mice and performed a detailed expression analysis for Eya2lacZ at different developmental stages to trace Eya2lacZ -positive cells in Eya2-null mice. We describe that Eya2 is not only expressed in cranial sensory and dorsal root ganglia, retina and olfactory epithelium, and somites as previously reported, but also Eya2 is specifically detected in other organs during mouse development. RESULTS: We found that Eya2 is expressed in ocular and trochlear motor neurons. In the inner ear, Eya2lacZ is specifically expressed in differentiating hair cells in both vestibular and cochlear sensory epithelia of the inner ear and Eya2-/- or Eya2lacZ/lacZ mice displayed mild hearing loss. Furthermore, we detected Eya2 expression during both salivary gland and thymus development and Eya2-null mice had a smaller thymus. CONCLUSIONS: As Eya2 is coexpressed with other members of the Eya family genes, these results together highlight that Eya2 as a potential regulator may act synergistically with other Eya genes to regulate the differentiation of the inner ear sensory hair cells and the formation of the salivary gland and thymus.


Asunto(s)
Oído Interno/metabolismo , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Pérdida Auditiva/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Diferenciación Celular/fisiología , Oído Interno/embriología , Pérdida Auditiva/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Tirosina Fosfatasas/genética
11.
Neurochem Res ; 46(10): 2525-2537, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33523395

RESUMEN

Astroglia are neural cells, heterogeneous in form and function, which act as supportive elements of the central nervous system; astrocytes contribute to all aspects of neural functions in health and disease. Through their highly ramified processes, astrocytes form close physical contacts with synapses and blood vessels, and are integrated into functional syncytia by gap junctions. Astrocytes interact among themselves and with other cells types (e.g., neurons, microglia, blood vessel cells) by an elaborate repertoire of chemical messengers and receptors; astrocytes also influence neural plasticity and synaptic transmission through maintaining homeostasis of neurotransmitters, K+ buffering, synaptic isolation and control over synaptogenesis and synaptic elimination. Satellite glial cells (SGCs) are the most abundant glial cells in sensory ganglia, and are believed to play major roles in sensory functions, but so far research into SGCs attracted relatively little attention. In this review we compare SGCs to astrocytes with the purpose of using the vast knowledge on astrocytes to explore new aspects of SGCs. We survey the main properties of these two cells types and highlight similarities and differences between them. We conclude that despite the much greater diversity in morphology and signaling mechanisms of astrocytes, there are some parallels between them and SGCs. Both types serve as boundary cells, separating different compartments in the nervous system, but much more needs to be learned on this aspect of SGCs. Astrocytes and SGCs employ chemical messengers and calcium waves for intercellular signaling, but their significance is still poorly understood for both cell types. Both types undergo major changes under pathological conditions, which have a protective function, but an also contribute to disease, and chronic pain in particular. The knowledge obtained on astrocytes is likely to benefit future research on SGCs.


Asunto(s)
Astrocitos/clasificación , Astrocitos/fisiología , Animales , Astrocitos/citología , Astrocitos/patología , Señalización del Calcio/fisiología , Extensiones de la Superficie Celular/fisiología , Uniones Comunicantes/fisiología , Humanos , Enfermedades del Sistema Nervioso/patología , Enfermedades del Sistema Nervioso/fisiopatología
12.
Methods Mol Biol ; 2241: 161-181, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33486736

RESUMEN

Eosinophils affect nerve structure and function in organs such as lungs and skin, which contributes to disease pathogenesis. We have developed methods for culturing primary sensory and parasympathetic neurons in multiple species and have refined these techniques for coculture with eosinophils. Eosinophil-nerve coculture has been an essential tool for testing interactions between these cell types. Here we describe methods for coculturing primary parasympathetic ganglia, vagal sensory nerves, and dorsal root sensory nerves with eosinophils.


Asunto(s)
Técnicas de Cocultivo/métodos , Eosinófilos/metabolismo , Neuronas/metabolismo , Animales , Técnicas de Cultivo de Célula/métodos , Eosinófilos/fisiología , Ganglios Parasimpáticos/metabolismo , Ganglios Espinales/metabolismo , Cobayas , Humanos , Tejido Nervioso/metabolismo , Neuronas/fisiología , Sistema Nervioso Parasimpático/metabolismo , Células Receptoras Sensoriales/metabolismo , Tráquea/citología , Nervio Vago/metabolismo
13.
Front Pain Res (Lausanne) ; 2: 646068, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35295432

RESUMEN

Chronic pain is known to be caused by sensitization within the pain circuits. An imbalance occurs between excitatory and inhibitory transmission that enables this sensitization to form. In addition to neurons, the contribution of central glia, especially astrocytes and microglia, to the pathogenesis of pain induction and maintenance has been identified. This has led to the targeting of astrogliosis and microgliosis to restore the normal functions of astrocytes and microglia to help reverse chronic pain. Gliosis is broadly defined as a reactive response of glial cells in response to insults to the central nervous system (CNS). The role of glia in the peripheral nervous system (PNS) has been less investigated. Accumulating evidence, however, points to the contribution of satellite glial cells (SGCs) to chronic pain. Hence, understanding the potential role of these cells and their interaction with sensory neurons has become important for identifying the mechanisms underlying pain signaling. This would, in turn, provide future therapeutic options to target pain. Here, a viewpoint will be presented regarding potential future directions in pain research, with a focus on SGCs to trigger further research. Promising avenues and new directions include the potential use of cell lines, cell live imaging, computational analysis, 3D tissue prints and new markers, investigation of glia-glia and macrophage-glia interactions, the time course of glial activation under acute and chronic pathological pain compared with spontaneous pain, pharmacological and non-pharmacological responses of glia, and potential restoration of normal function of glia considering sex-related differences.

14.
Front Cell Neurosci ; 14: 612982, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33362476

RESUMEN

Despite the widespread study of how injured nerves contribute to chronic pain, there are still major gaps in our understanding of pain mechanisms. This is particularly true of pain resulting from nerve injury, or neuropathic pain, wherein tactile or thermal stimuli cause painful responses that are particularly difficult to treat with existing therapies. Curiously, this stimulus-driven pain relies upon intact, uninjured sensory neurons that transmit the signals that are ultimately sensed as painful. Studies that interrogate uninjured neurons in search of cell-specific mechanisms have shown that nerve injury alters intact, uninjured neurons resulting in an activity that drives stimulus-evoked pain. This review of neuropathic pain mechanisms summarizes cell-type-specific pathology of uninjured sensory neurons and the sensory ganglia that house their cell bodies. Uninjured neurons have demonstrated a wide range of molecular and neurophysiologic changes, many of which are distinct from those detected in injured neurons. These intriguing findings include expression of pain-associated molecules, neurophysiological changes that underlie increased excitability, and evidence that intercellular signaling within sensory ganglia alters uninjured neurons. In addition to well-supported findings, this review also discusses potential mechanisms that remain poorly understood in the context of nerve injury. This review highlights key questions that will advance our understanding of the plasticity of sensory neuron subpopulations and clarify the role of uninjured neurons in developing anti-pain therapies.

15.
Front Cell Dev Biol ; 8: 587699, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33195244

RESUMEN

Sensory fibers of the peripheral nervous system carry sensation from specific sense structures or use different tissues and organs as receptive fields, and convey this information to the central nervous system. In the head of vertebrates, each cranial sensory ganglia and associated nerves perform specific functions. Sensory ganglia are composed of different types of specialized neurons in which two broad categories can be distinguished, somatosensory neurons relaying all sensations that are felt and visceral sensory neurons sensing the internal milieu and controlling body homeostasis. While in the trunk somatosensory neurons composing the dorsal root ganglia are derived exclusively from neural crest cells, somato- and visceral sensory neurons of cranial sensory ganglia have a dual origin, with contributions from both neural crest and placodes. As most studies on sensory neurogenesis have focused on dorsal root ganglia, our understanding of the molecular mechanisms underlying the embryonic development of the different cranial sensory ganglia remains today rudimentary. However, using single-cell RNA sequencing, recent studies have made significant advances in the characterization of the neuronal diversity of most sensory ganglia. Here we summarize the general anatomy, function and neuronal diversity of cranial sensory ganglia. We then provide an overview of our current knowledge of the transcriptional networks controlling neurogenesis and neuronal diversification in the developing sensory system, focusing on cranial sensory ganglia, highlighting specific aspects of their development and comparing it to that of trunk sensory ganglia.

16.
Epigenet Insights ; 13: 2516865720938677, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32974606

RESUMEN

Epigenetic modulation by DNA methylation is associated with aberrant gene expression in sensory neurons, which consequently leads to pathological pain responses. In this study, we sought to investigate whether peripheral inflammation alters global DNA methylation in trigeminal ganglia (TG) and results in abnormal expression of pro-nociceptive genes. Our results show that peripheral inflammation remotely reduced the level of global DNA methylation in rat TG with a concurrent reduction in DNMT1 and DNMT3a expression. Using unbiased steps, we selected the following pro-nociceptive candidate genes that are potentially regulated by DNA methylation: TRPV1, TRPA1, P2X3, and PIEZO2. Inhibition of DNMT with 5-Aza-dC in dissociated TG cells produced dose-dependent upregulation of TRPV1, TRPA1, and P2X3. Systemic treatment of animals with 5-Aza-dC significantly increased the expression of TRPV1, TRPA1, and PIEZO2 in TG. Furthermore, the overexpression of DNMT3a, as delivered by a lentiviral vector, significantly downregulated TRPV1 and PIEZO2 expression and also reliably decreased TRPA1 and P2X3 transcripts. MeDIP revealed that this overexpression also significantly enhanced methylation of CGIs associated with TRPV1 and TRPA1. In addition, bisulfite sequencing data indicated that the CGI associated with TRPA1 was methylated in a pattern catalyzed by DNMT3a. Taken together, our results show that all 4 pro-nociceptive genes are subject to epigenetic modulation via DNA methylation, likely via DNMT3a under inflammatory conditions. These findings provide the first evidence for the functional importance of DNA methylation as an epigenetic factor in the transcription of pro-nociceptive genes in TG that are implicated in pathological orofacial pain responses.

17.
Curr Top Dev Biol ; 139: 127-167, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32450959

RESUMEN

Sensory placodes and neural crest cells are among the key cell populations that facilitated the emergence and diversification of vertebrates throughout evolution. Together, they generate the sensory nervous system in the head: both form the cranial sensory ganglia, while placodal cells make major contributions to the sense organs-the eye, ear and olfactory epithelium. Both are instrumental for integrating craniofacial organs and have been key to drive the concentration of sensory structures in the vertebrate head allowing the emergence of active and predatory life forms. Whereas the gene regulatory networks that control neural crest cell development have been studied extensively, the signals and downstream transcriptional events that regulate placode formation and diversity are only beginning to be uncovered. Both cell populations are derived from the embryonic ectoderm, which also generates the central nervous system and the epidermis, and recent evidence suggests that their initial specification involves a common molecular mechanism before definitive neural, neural crest and placodal lineages are established. In this review, we will first discuss the transcriptional networks that pattern the embryonic ectoderm and establish these three cell fates with emphasis on sensory placodes. Second, we will focus on how sensory placode precursors diversify using the specification of otic-epibranchial progenitors and their segregation as an example.


Asunto(s)
Diferenciación Celular/genética , Ectodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Sistema Nervioso Periférico/metabolismo , Vertebrados/genética , Animales , Ectodermo/citología , Ectodermo/embriología , Cabeza/embriología , Humanos , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Sistema Nervioso Periférico/citología , Sistema Nervioso Periférico/embriología , Vertebrados/clasificación , Vertebrados/embriología
18.
Neurosci Biobehav Rev ; 108: 393-399, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31785264

RESUMEN

Neuropathic pain (NP) develops because of damage to the peripheral or central nervous system. It results in the hyperalgesia and allodynia. In the recent years, various researchers have studied the involvement of neuro-immune system in causing persistence of pain. The absence of synaptic contacts in the sensory ganglion makes them distinctive in terms of pain related signalling. In sensory ganglia, the neurotransmitters or the other modulators such as inflammatory substances produced by the ganglion cells, because of an injury, are responsible for the cross-excitation between neurons and neuron-glial interaction, thus affecting chemical transmission. This chemical transmission is considered mainly responsible for the chronicity and the persistent nature of neuropathic pain. This review examines the pain signalling due to neurotransmitter or cytokine release within the sensory ganglia. The specific areas focused on include: 1) the role of neurotransmitters released from the somata of sensory neurons in pain, 2) neuron-glia interaction and 3) role of cytokines in neuromodulation and pain.


Asunto(s)
Citocinas/metabolismo , Ganglios Sensoriales/metabolismo , Neuralgia/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Neurotransmisores/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Neuralgia/inmunología
19.
Neurosci Lett ; 703: 132-138, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-30904573

RESUMEN

Many trigeminal neuropathic pain patients suffer severe chronic pain. The neuropathic pain might be related with cross-excitation of the neighboring neurons and satellite glial cells (SGCs) in the sensory ganglia and increasing the pain signals from the peripheral tissue to the central nervous system. We induced trigeminal neuropathic pain by infraorbital nerve constriction injury (IONC) in Sprague-Dawley rats. We tested cytokine (CXCL2 and IL-10) levels in trigeminal ganglia (TGs) after trigeminal neuropathic pain induction, and the effect of direct injection of the anti-CXCL2 and recombinant IL-10 into TG. We found that IONC induced pain behavior. Additionally, IONC induced satellite glial cell activation in TG and cytokine levels of TGs were changed after IONC. CXCL2 levels increased on day 1 of neuropathic pain induction and decreased gradually, with IL-10 levels showing the opposite trend. Recombinant IL-10 or anti-CXCL2 injection into TG decreased pain behavior. Our results show that IL-10 or anti-CXCL2 are therapy options for neuropathic pain.


Asunto(s)
Quimiocina CXCL2/metabolismo , Interleucina-10/metabolismo , Neuralgia/metabolismo , Ganglio del Trigémino/metabolismo , Animales , Anticuerpos/farmacología , Quimiocina CXCL2/inmunología , Constricción Patológica , Interleucina-10/farmacología , Masculino , Neuralgia/fisiopatología , Dimensión del Dolor , Traumatismos de los Nervios Periféricos/fisiopatología , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología
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