Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Cells ; 13(7)2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38607005

RESUMO

Satellite glial cells (SGCs) are the main type of glial cells in sensory ganglia. Animal studies have shown that these cells play essential roles in both normal and disease states. In a large number of pain models, SGCs were activated and contributed to the pain behavior. Much less is known about SGCs in humans, but there is emerging recognition that SGCs in humans are altered in a variety of clinical states. The available data show that human SGCs share some essential features with SGCs in rodents, but many differences do exist. SGCs in DRG from patients suffering from common painful diseases, such as rheumatoid arthritis and fibromyalgia, may contribute to the pain phenotype. It was found that immunoglobulins G (IgG) from fibromyalgia patients can induce pain-like behavior in mice. Moreover, these IgGs bind preferentially to SGCs and activate them, which can sensitize the sensory neurons, causing nociception. In other human diseases, the evidence is not as direct as in fibromyalgia, but it has been found that an antibody from a patient with rheumatoid arthritis binds to mouse SGCs, which leads to the release of pronociceptive factors from them. Herpes zoster is another painful disease, and it appears that the zoster virus resides in SGCs, which acquire an abnormal morphology and may participate in the infection and pain generation. More work needs to be undertaken on SGCs in humans, and this review points to several promising avenues for better understanding disease mechanisms and developing effective pain therapies.


Assuntos
Artrite Reumatoide , Fibromialgia , Humanos , Camundongos , Animais , Neuroglia/fisiologia , Dor , Células Receptoras Sensoriais
2.
Glia ; 72(6): 1054-1066, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38450799

RESUMO

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.


Assuntos
Neuroglia , Neurônios , Animais , Camundongos , Neuroglia/metabolismo , Gânglios Sensitivos , Gânglios Espinais , Glutamatos/metabolismo , Trifosfato de Adenosina/metabolismo , Células Satélites Perineuronais/metabolismo
3.
Int J Mol Sci ; 24(3)2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36769006

RESUMO

The effects of aging on the nervous system are well documented. However, most previous studies on this topic were performed on the central nervous system. The present study was carried out on the dorsal root ganglia (DRGs) of mice, and focused on age-related changes in DRG neurons and satellite glial cells (SGCs). Intracellular electrodes were used for dye injection to examine the gap junction-mediated coupling between neurons and SGCs, and for intracellular electrical recordings from the neurons. Tactile sensitivity was assessed with von Frey hairs. We found that 3-23% of DRG neurons were dye-coupled to SGCs surrounding neighboring neurons in 8-24-month (Mo)-old mice, whereas in young adult (3 Mo) mice, the figure was 0%. The threshold current for firing an action potential in sensory neurons was significantly lower in DRGs from 12 Mo mice compared with those from 3 Mo mice. The percentage of neurons with spontaneous subthreshold membrane potential oscillation was greater by two-fold in 12 Mo mice. The withdrawal threshold was lower by 22% in 12 Mo mice compared with 3 Mo ones. These results show that in the aged mice, a proportion of DRG neurons is coupled to SGCs, and that the membrane excitability of the DRG neurons increases with age. We propose that augmented neuron-SGC communications via gap junctions are caused by low-grade inflammation associated with aging, and this may contribute to pain behavior.


Assuntos
Gânglios Espinais , Neuroglia , Camundongos , Animais , Potenciais da Membrana , Células Receptoras Sensoriais , Camundongos Endogâmicos BALB C
4.
Cells ; 11(15)2022 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-35892578

RESUMO

The purinergic system plays an important role in pain transmission. Recent studies have suggested that activation of P2-purinergic receptors (P2Rs) may be involved in neuron-satellite glial cell (SGC) interactions in the dorsal root ganglia (DRG), but the details remain unclear. In DRG, P2X7R is selectively expressed in SGCs, which closely surround neurons, and is highly sensitive to 3'-O-(4-Benzoyl) benzoyl-ATP (BzATP). Using calcium imaging in intact mice to survey a large number of DRG neurons and SGCs, we examined how intra-ganglionic purinergic signaling initiated by BzATP affects neuronal activities in vivo. We developed GFAP-GCaMP6s and Pirt-GCaMP6s mice to express the genetically encoded calcium indicator GGCaM6s in SGCs and DRG neurons, respectively. The application of BzATP to the ganglion induced concentration-dependent activation of SGCs in GFAP-GCaMP6s mice. In Pirt-GCaMP6s mice, BzATP initially activated more large-size neurons than small-size ones. Both glial and neuronal responses to BzATP were blocked by A438079, a P2X7R-selective antagonist. Moreover, blockers to pannexin1 channels (probenecid) and P2X3R (A317491) also reduced the actions of BzATP, suggesting that P2X7R stimulation may induce the opening of pannexin1 channels, leading to paracrine ATP release, which could further excite neurons by acting on P2X3Rs. Importantly, BzATP increased the responses of small-size DRG neurons and wide-dynamic range spinal neurons to subsequent peripheral stimuli. Our findings suggest that intra-ganglionic purinergic signaling initiated by P2X7R activation could trigger SGC-neuron interaction in vivo and increase DRG neuron excitability.


Assuntos
Cálcio , Gânglios Espinais , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Camundongos , Neuroglia , Neurônios/fisiologia
5.
Neurosci Lett ; 778: 136616, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35381320

RESUMO

Emerging research indicates that physical activity can ameliorate chronic pain, but the underlying mechanisms are still largely obscure. In particular, little is known on the mechanisms behind exercise-induced analgesia in the setting of inflammatory pain. In our previous studies on systemic inflammation in mice using lipopolysaccharide (LPS) administration, we characterized satellite glial cells (SGCs) and neurons in dorsal root ganglia (DRG). We found that a week post-LPS injection, the sensitivity to mechanical stimulation was lowered, SGCs were activated and coupling among SGCs increased 3 to 4.5-fold. In the present work, we examined the effects of exercise (free wheel running) on tactile sensitivity and on pathological changes in mouse DRG in the LPS model. We found that exercise prevented tactile hypersensitivity, and also reversed the cellular changes in the DRG induced by LPS that were listed above. We propose that the analgesic effect of exercise is at least partly mediated by reversing the pathological changes in SGCs.


Assuntos
Gânglios Espinais , Lipopolissacarídeos , Animais , Gânglios Espinais/patologia , Junções Comunicantes/fisiologia , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Atividade Motora , Neuroglia/patologia , Dor/patologia
6.
Cells ; 11(5)2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35269508

RESUMO

Itch (pruritus) is a common chronic condition with a lifetime prevalence of over 20%. The mechanisms underlying itch are poorly understood, and its therapy is difficult. There is recent evidence that following nerve injury or inflammation, intercellular communications in sensory ganglia are augmented, which may lead to abnormal neuronal activity, and hence to pain, but there is no information whether such changes take place in an itch model. We studied changes in neurons and satellite glial cells (SGCs) in trigeminal ganglia in an itch model in mice using repeated applications of 2,4,6-trinitro-1-chlorobenzene (TNCB) to the external ear over a period of 11 days. Treated mice showed augmented scratching behavior as compared with controls during the application period and for several days afterwards. Immunostaining for the activation marker glial fibrillary acidic protein in SGCs was greater by about 35% after TNCB application, and gap junction-mediated coupling between neurons increased from about 2% to 13%. The injection of gap junction blockers reduced scratching behavior, suggesting that gap junctions contribute to itch. Calcium imaging studies showed increased responses of SGCs to the pain (and presumed itch) mediator ATP. We conclude that changes in both neurons and SGCs in sensory ganglia may play a role in itch.


Assuntos
Neuroglia , Gânglio Trigeminal , Animais , Camundongos , Neuroglia/metabolismo , Neurônios/metabolismo , Dor/metabolismo , Prurido , Gânglio Trigeminal/metabolismo
7.
Cells ; 11(3)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35159321

RESUMO

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.


Assuntos
Dor Crônica , Células Satélites Perineuronais , Dor Crônica/metabolismo , Gânglios Sensitivos , Humanos , Inflamação/metabolismo , Neuroglia/metabolismo , Células Satélites Perineuronais/metabolismo
8.
Pain ; 163(8): 1636-1647, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35027518

RESUMO

ABSTRACT: Primary sensory neurons in dorsal root ganglia (DRG) are wrapped by satellite glial cells (SGCs), and neuron-SGC interaction may affect somatosensation, especially nociceptive transmission. P2-purinergic receptors (P2Rs) are key elements in the two-way interactions between DRG neurons and SGCs. However, because the cell types are in such close proximity, conventional approaches such as in vitro culture and electrophysiologic recordings are not adequate to investigate the physiologically relevant responses of these cells at a population level. Here, we performed in vivo calcium imaging to survey the activation of hundreds of DRG neurons in Pirt-GCaMP6s mice and to assess SGC activation in GFAP-GCaMP6s mice in situ. By combining pharmacologic and electrophysiologic techniques, we investigated how ganglionic purinergic signaling initiated by α,ß-methyleneadenosine 5'-triphosphate (α,ß-MeATP) modulates neuronal activity and excitability at a population level. We found that α,ß-MeATP induced robust activation of small neurons-likely nociceptors-through activation of P2X3R. Large neurons, which are likely non-nociceptive, were also activated by α,ß-MeATP, but with a delay. Blocking pannexin 1 channels attenuated the late phase response of DRG neurons, indicating that P2R stimulation may subsequently induce paracrine ATP release, which could further activate cells in the ganglion. Moreover, ganglionic α,ß-MeATP treatment in vivo sensitized small neurons and enhanced responses of spinal wide-dynamic-range neurons to subsequent C-fiber inputs, suggesting that modulation via ganglionic P2R signaling could significantly affect nociceptive neuron excitability and pain transmission. Therefore, targeting functional P2Rs within ganglia may represent an important new strategy for pain modulation.


Assuntos
Gânglios Espinais , Neuroglia , Animais , Humanos , Camundongos , Neurônios/metabolismo , Dor/metabolismo , Transdução de Sinais
9.
J Physiol ; 599(17): 4225-4226, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34031884
10.
Sci Adv ; 7(11)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33692101

RESUMO

No disease-modifying therapy is currently available for Parkinson's disease (PD), the second most common neurodegenerative disease. The long nonmotor prodromal phase of PD is a window of opportunity for early detection and intervention. However, we lack the pathophysiological understanding to develop selective biomarkers and interventions. By using a mutant α-synuclein selective-overexpression mouse model of prodromal PD, we identified a cell-autonomous selective Kv4 channelopathy in dorsal motor nucleus of the vagus (DMV) neurons. This functional remodeling of intact DMV neurons leads to impaired pacemaker function in vitro and in vivo, which, in turn, reduces gastrointestinal motility, a common early symptom of prodromal PD. We identify a chain of events from α-synuclein via a biophysical dysfunction of a specific neuronal population to a clinically relevant prodromal symptom. These findings will facilitate the rational design of clinical biomarkers to identify people at risk for developing PD.


Assuntos
Canalopatias , Doenças Neurodegenerativas , Doença de Parkinson , Animais , Humanos , Camundongos , Neurônios Motores , Doença de Parkinson/etiologia , alfa-Sinucleína/genética
11.
Brain Res ; 1760: 147384, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33631206

RESUMO

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.


Assuntos
Comunicação Celular/fisiologia , Gânglios Espinais/metabolismo , Neurônios/metabolismo , Células Satélites Perineuronais/metabolismo , Gânglio Trigeminal/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neurotransmissores/metabolismo , Óxido Nítrico/metabolismo
12.
Neurochem Res ; 46(10): 2525-2537, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33523395

RESUMO

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.


Assuntos
Astrócitos/classificação , Astrócitos/fisiologia , Animais , Astrócitos/citologia , Astrócitos/patologia , Sinalização do Cálcio/fisiologia , Extensões da Superfície Celular/fisiologia , Junções Comunicantes/fisiologia , Humanos , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/fisiopatologia
13.
Nat Rev Neurosci ; 21(12): 732, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33093636

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

14.
Nat Rev Neurosci ; 21(9): 485-498, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32699292

RESUMO

Satellite glial cells (SGCs) closely envelop cell bodies of neurons in sensory, sympathetic and parasympathetic ganglia. This unique organization is not found elsewhere in the nervous system. SGCs in sensory ganglia are activated by numerous types of nerve injury and inflammation. The activation includes upregulation of glial fibrillary acidic protein, stronger gap junction-mediated SGC-SGC and neuron-SGC coupling, increased sensitivity to ATP, downregulation of Kir4.1 potassium channels and increased cytokine synthesis and release. There is evidence that these changes in SGCs contribute to chronic pain by augmenting neuronal activity and that these changes are consistent in various rodent pain models and likely also in human pain. Therefore, understanding these changes and the resulting abnormal interactions of SGCs with sensory neurons could provide a mechanistic approach that might be exploited therapeutically in alleviation and prevention of pain. We describe how SGCs are altered in rodent models of four common types of pain: systemic inflammation (sickness behaviour), post-surgical pain, diabetic neuropathic pain and post-herpetic pain.


Assuntos
Dor Crônica/fisiopatologia , Gânglios Autônomos/fisiopatologia , Gânglios Sensitivos/fisiopatologia , Células Satélites Perineuronais/fisiologia , Animais , Humanos
15.
Auton Neurosci ; 221: 102584, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31494528

RESUMO

Neurons in sensory, sympathetic, and parasympathetic ganglia are surrounded by satellite glial cell (SGCs). There is little information on the effects of nerve damage on SGCs in autonomic ganglia. We studied the consequences of damage to sympathetic nerve terminals by 6-hydroxydopamine (6-OHDA) on SGCs in the mouse superior cervical ganglia (Sup-CG). Immunostaining revealed that at 1-30 d post-6-OHDA injection, SGCs in Sup-CG were activated, as assayed by upregulation of glial fibrillary acidic protein. Intracellular labeling showed that dye coupling between SGCs around different neurons increased 4-6-fold 1-14 d after 6-OHDA injection. Behavioral testing 1-7 d post-6-OHDA showed that withdrawal threshold to tactile stimulation of the hind paws was reduced by 65-85%, consistent with hypersensitivity. A single intraperitoneal injection of the gap junction blocker carbenoxolone restored normal tactile thresholds in 6-OHDA-treated mice, suggesting a contribution of SGC gap junctions to pain. Using calcium imaging we found that after 6-OHDA treatment responses of SGCs to ATP were increased by about 30% compared with controls, but responses to ACh were reduced by 48%. The same experiments for SGCs in trigeminal ganglia from 6-OHDA injected mice showed no difference from controls, confirming that 6-OHDA acted selectively on sympathetic nerves. However, systemic inflammation induced by lipopolysaccharide did not affect SGCs of Sup-CG, but did influence SGCs in trigeminal ganglia in the same manner as 6-OHDA did on SGCs in Sup-CG. We conclude that even though SGCs in sympathetic and sensory ganglia are morphologically similar, they are quite different functionally, particularly after damage.


Assuntos
Células Satélites Perineuronais/fisiologia , Gânglio Cervical Superior/patologia , Sistema Nervoso Simpático/efeitos dos fármacos , Acetilcolina/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Sinalização do Cálcio , Carbenoxolona/farmacologia , Comunicação Celular/efeitos dos fármacos , Feminino , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/fisiologia , Glutamato-Amônia Ligase/biossíntese , Glutamato-Amônia Ligase/genética , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neuralgia/fisiopatologia , Oxidopamina/toxicidade , Limiar da Dor/fisiologia , Células Satélites Perineuronais/efeitos dos fármacos , Receptor 4 Toll-Like/biossíntese , Receptor 4 Toll-Like/genética , Gânglio Trigeminal/patologia
16.
Glia ; 67(5): 791-801, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30715764

RESUMO

Peripheral sensory ganglia contain the somata of neurons mediating mechanical, thermal, and painful sensations from somatic, visceral, and oro-facial organs. Each neuronal cell body is closely surrounded by satellite glial cells (SGCs) that have properties and functions similar to those of central astrocytes, including expression of gap junction proteins and functional dye coupling. As shown in other pain models, after systemic pain induction by intra-peritoneal injection of lipopolysaccharide, dye coupling among SGCs in intact trigeminal ganglion was enhanced. Moreover, neuron-neuron and neuron-SGC coupling was also detected. To verify the presence of gap junction-mediated coupling between SGCs and sensory neurons, we performed dual whole cell patch clamp recordings from both freshly isolated and short term cultured cell pairs dissociated from mouse trigeminal ganglia. Bidirectional gap junction mediated electrical responses were frequently recorded between SGCs, between neurons and between neurons and SGCs. Polarization of SGC altered neuronal excitability, providing evidence that gap junction-mediated interactions between neurons and glia within sensory ganglia may contribute to integration of peripheral sensory responses, and to the modulation and coordinaton of neuronal activity.


Assuntos
Junções Comunicantes/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia , Transmissão Sináptica/fisiologia , Gânglio Trigeminal/citologia , Animais , Compostos de Boro/farmacologia , Carbenoxolona/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Ácido Flufenâmico/farmacologia , Junções Comunicantes/efeitos dos fármacos , Heptanol/farmacologia , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/patologia , Isoquinolinas/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Probenecid/farmacologia , Transmissão Sináptica/efeitos dos fármacos
17.
Glia ; 67(7): 1296-1307, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30801760

RESUMO

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.


Assuntos
Comunicação Celular/fisiologia , Gânglios Espinais/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Óxido Nítrico/biossíntese , Células Satélites Perineuronais/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Dinitrofluorbenzeno/análogos & derivados , Dinitrofluorbenzeno/farmacologia , Feminino , Gânglios Espinais/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Células Satélites Perineuronais/efeitos dos fármacos
18.
Neurosci Lett ; 695: 46-52, 2019 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28647288

RESUMO

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.


Assuntos
Conexinas/metabolismo , Junções Comunicantes/metabolismo , Dor/metabolismo , Animais , Gânglios Sensitivos/metabolismo , Gânglios Sensitivos/patologia , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Neuralgia/metabolismo , Neuralgia/patologia , Neuroglia/metabolismo , Neuroglia/patologia , Dor/patologia , Células Satélites Perineuronais/metabolismo , Células Satélites Perineuronais/patologia , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/patologia
19.
Neurosci Lett ; 671: 19-24, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29391220

RESUMO

Satellite glial cells (SGCs) surround the neurons in sympathetic ganglia and are believed to make important contributions to the function of the ganglia under normal and pathological conditions. It has been proposed that SGCs communicate chemically with the neurons, but little is known about their pharmacological properties and there is no information on whether they respond to acetylcholine (ACh), which is the major neurotransmitter in these ganglia. We used calcium imaging to examine responses of SGCs in the mouse superior cervical ganglion to ACh. The SGCs responded to ACh (0.01-2 mM) with an elevation of intracellular Ca2+, which appeared to be due to direct action on these cells, as the response persisted in the presence of the nerve blocker tetrodotoxin (1 µM). The response was largely inhibited by atropine, indicating an action on muscarinic ACh receptors. In contrast to this, sensory ganglia (nodose and trigeminal) were not sensitive to ACh. Incubation of the ganglia in ACh (0.5 or 1 mM) increased the expression of glial fibrillay acidic protein, which is a marker for glial activation. Such incubation also increased the electrical coupling of SGCs, which is known to occur in sensory ganglia following injury. We conclude that SGCs in the superior cervical ganglia display muscarinic ACh receptors, which enable them to communicate chemically with the sympathetic neurons.


Assuntos
Acetilcolina/farmacologia , Colinérgicos/farmacologia , Células Satélites Perineuronais/efeitos dos fármacos , Gânglio Cervical Superior/efeitos dos fármacos , Animais , Atropina/farmacologia , Cálcio/metabolismo , Camundongos , Antagonistas Muscarínicos/farmacologia , Células Satélites Perineuronais/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Gânglio Cervical Superior/metabolismo , Tetrodotoxina/farmacologia
20.
Neuropeptides ; 63: 37-42, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28342550

RESUMO

Endothelins (ET) are a family of highly active neuropeptides with manifold influences via ET receptors (ETR) in both the peripheral and central nervous systems. We have shown previously that satellite glial cells (SGCs) in mouse trigeminal ganglia (TG) are extremely sensitive to ET-1 in evoking [Ca2+]in increase, apparently via ETBR activation, but there is no functional information on ETR in SGCs of other peripheral ganglia. Here we tested the effects of ET-1 on SGCs in nodose ganglia (NG), which is sensory, and superior cervical ganglia (Sup-CG), which is part of the sympathetic nervous system, and further investigated the influence of ET-1 on SGCs in TG. Using calcium imaging we found that SGCs in intact, freshly isolated NG and Sup-CG are highly sensitive to ET-1, with threshold concentration at 0.1nM. Our results showed that [Ca2+]in elevation in response to ET-1 was partially due to Ca2+ influx from the extracellular space and partially to Ca2+ release from intracellular stores. Using receptor selective ETR agonists and antagonists, we found that the responses were mediated by mixed ETAR/ETBR in SGCs of NG and predominantly by ETBR in SGCs of Sup-CG. By employing intracellular dye injection we examined coupling among SGCs around different neurons in the presence of 5nM ET-1 and observed coupling inhibition in all the three ganglion types. In summary, our work showed that SGCs in mouse sensory and sympathetic ganglia are highly sensitive to ET-1 and that this peptide markedly reduces SGCs coupling. We conclude that ET-1, which may participate in neuron-glia communications, has similar functions in wide range of peripheral ganglia.


Assuntos
Cálcio/metabolismo , Endotelina-1/farmacologia , Gânglios Simpáticos/efeitos dos fármacos , Células Satélites Perineuronais/efeitos dos fármacos , Gânglio Trigeminal/efeitos dos fármacos , Animais , Antagonistas dos Receptores de Endotelina/farmacologia , Gânglios Simpáticos/metabolismo , Camundongos , Células Satélites Perineuronais/metabolismo , Gânglio Trigeminal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA