Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 4 de 4
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Brain Behav Immun ; 117: 224-241, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38244946

RÉSUMÉ

Glial activation and dysregulation of adenosine triphosphate (ATP)/adenosine are involved in the neuropathology of several neuropsychiatric illnesses. The ventral hippocampus (vHPC) has attracted considerable attention in relation to its role in emotional regulation. However, it is not yet clear how vHPC glia and their derived adenosine regulate the anxiodepressive-like consequences of chronic pain. Here, we report that chronic cheek pain elevates vHPC extracellular ATP/adenosine in a mouse model resembling trigeminal neuralgia (rTN), which mediates pain-related anxiodepression, through a mechanism that involves synergistic effects of astrocytes and microglia. We found that rTN resulted in robust activation of astrocytes and microglia in the CA1 area of the vHPC (vCA1). Genetic or pharmacological inhibition of astrocytes and connexin 43, a hemichannel mainly distributed in astrocytes, completely attenuated rTN-induced extracellular ATP/adenosine elevation and anxiodepressive-like behaviors. Moreover, inhibiting microglia and CD39, an enzyme primarily expressed in microglia that degrades ATP into adenosine, significantly suppressed the increase in extracellular adenosine and anxiodepressive-like behaviors. Blockade of the adenosine A2A receptor (A2AR) alleviated rTN-induced anxiodepressive-like behaviors. Furthermore, interleukin (IL)-17A, a pro-inflammatory cytokine probably released by activated microglia, markedly increased intracellular calcium in vCA1 astrocytes and triggered ATP/adenosine release. The astrocytic metabolic inhibitor fluorocitrate and the CD39 inhibitor ARL 67156, attenuated IL-17A-induced increases in extracellular ATP and adenosine, respectively. In addition, astrocytes, microglia, CD39, and A2AR inhibitors all reversed rTN-induced hyperexcitability of pyramidal neurons in the vCA1. Taken together, these findings suggest that activation of astrocytes and microglia in the vCA1 increases extracellular adenosine, which leads to pain-related anxiodepression via A2AR activation. Approaches targeting astrocytes, microglia, and adenosine signaling may serve as novel therapies for pain-related anxiety and depression.


Sujet(s)
Douleur chronique , Névralgie essentielle du trijumeau , Animaux , Souris , Adénosine/pharmacologie , Adénosine triphosphate/pharmacologie , Modèles animaux de maladie humaine , Hippocampe , Microglie
2.
JCI Insight ; 5(20)2020 10 15.
Article de Anglais | MEDLINE | ID: mdl-32960817

RÉSUMÉ

Recently programmed death-ligand 1 (PD-L1) receptor PD-1 was found in dorsal root ganglion (DRG) neurons, and PD-L1 activates PD-1 to inhibit inflammatory and neuropathic pain by modulating neuronal excitability. However, the downstream signaling of PD-1 in sensory neurons remains unclear. Here, we show that PD-L1 activated Src homology 2 domain-containing tyrosine phosphatase-1 (SHP-1) to downregulate transient receptor potential vanilloid 1 (TRPV1) in DRG neurons and inhibit bone cancer pain in mice. Local injection of PD-L1 produced analgesia. PD-1 in DRG neurons colocalized with TRPV1 and SHP-1. PD-L1 induced the phosphorylation of SHP-1 in DRG TRPV1 neurons and inhibited TRPV1 currents. Loss of TRPV1 in mice abolished bone cancer-induced thermal hyperalgesia and PD-L1 analgesia. Conditioned deletion of SHP-1 in NaV1.8+ neurons aggravated bone cancer pain and diminished the inhibition of PD-L1 on TRPV1 currents and pain. Together, our findings suggest that PD-L1/PD-1 signaling suppresses bone cancer pain via inhibition of TRPV1 activity. Our results also suggest that SHP-1 in sensory neurons is an endogenous pain inhibitor and delays the development of bone cancer pain via suppressing TRPV1 function.


Sujet(s)
Antigène CD274/génétique , Douleur cancéreuse/génétique , Névralgie/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 6/génétique , Canaux cationiques TRPV/génétique , Analgésie/méthodes , Animaux , Tumeurs osseuses/complications , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Douleur cancéreuse/complications , Douleur cancéreuse/anatomopathologie , Modèles animaux de maladie humaine , Ganglions sensitifs des nerfs spinaux/métabolisme , Ganglions sensitifs des nerfs spinaux/anatomopathologie , Inflammation/complications , Inflammation/génétique , Inflammation/anatomopathologie , Mâle , Souris , Canal sodique voltage-dépendant NAV1.8/génétique , Névralgie/complications , Névralgie/anatomopathologie , Récepteur-1 de mort cellulaire programmée/génétique , Cellules réceptrices sensorielles/anatomopathologie
3.
Cell Rep ; 29(8): 2384-2397.e5, 2019 11 19.
Article de Anglais | MEDLINE | ID: mdl-31747607

RÉSUMÉ

The proinflammatory cytokine interleukin-17 (IL-17) is implicated in pain regulation. However, the synaptic mechanisms by which IL-17 regulates pain transmission are unknown. Here, we report that glia-produced IL-17 suppresses inhibitory synaptic transmission in the spinal cord pain circuit and drives chemotherapy-induced neuropathic pain. We find that IL-17 not only enhances excitatory postsynaptic currents (EPSCs) but also suppresses inhibitory postsynaptic synaptic currents (IPSCs) and GABA-induced currents in lamina IIo somatostatin-expressing neurons in mouse spinal cord slices. IL-17 mainly expresses in spinal cord astrocytes, and its receptor IL-17R is detected in somatostatin-expressing neurons. Selective knockdown of IL-17R in spinal somatostatin-expressing interneurons reduces paclitaxel-induced hypersensitivity. Overexpression of IL-17 in spinal astrocytes is sufficient to induce mechanical allodynia in naive animals. In dorsal root ganglia, IL-17R expression in nociceptive sensory neurons is sufficient and required for inducing neuronal hyperexcitability after paclitaxel. Together, our data show that IL-17/IL-17R mediate neuron-glial interactions and neuronal hyperexcitability in chemotherapy-induced peripheral neuropathy.


Sujet(s)
Interleukine-17/métabolisme , Névralgie/métabolisme , Transmission synaptique/physiologie , Animaux , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Potentiels post-synaptiques excitateurs/effets des médicaments et des substances chimiques , Potentiels post-synaptiques excitateurs/physiologie , Humains , Névralgie/physiopathologie , Névroglie/effets des médicaments et des substances chimiques , Névroglie/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neuropathies périphériques/métabolisme , Somatostatine/métabolisme , Transmission synaptique/effets des médicaments et des substances chimiques , Acide gamma-amino-butyrique/pharmacologie
4.
Sheng Li Xue Bao ; 68(3): 224-32, 2016 Jun 25.
Article de Chinois | MEDLINE | ID: mdl-27350194

RÉSUMÉ

Cancer pain is one of the most common symptoms in patients with late stage cancer. Lung, breast and prostate carcinoma are the most common causes of pain from osseous metastasis. P2X7 receptor (P2X7R) is one of the subtypes of ATP-gated purinergic ion channel family, predominately distributed in microglia in the spinal cord. Activation of P2X7Rs in the spinal dorsal horn has been associated with release of proinflammatory cytokines from glial cells, causing increased neuronal excitability and exaggerated nociception. Mounting evidence implies a critical role of P2X7R in inflammatory and neuropathic pain. However, whether P2X7R is involved in cancer pain remains controversial. Here we established a bone cancer pain model by injecting the Lewis lung carcinoma cells into the femur bone marrow cavity of C57BL/6J wild-type mice (C57 WT mice) and P2X7R knockout mice (P2rx7(-/-) mice) to explore the role of P2X7R in bone cancer pain. Following intrafemur carcinoma inoculation, robust mechanical allodynia and thermal hyperalgesia in C57 WT mice were developed on day 7 and 14, respectively, and persisted for at least 28 days in the ipsilateral hindpaw of the affected limb. CatWalk gait analysis showed significant decreases in the print area and stand phase, and a significant increase in swing phase in the ipsilateral hindpaw on day 21 and 28 after carcinoma cells inoculation. Histopathological sections (hematoxylin and eosin stain) showed that the bone marrow of the affected femur was largely replaced by invading tumor cells, and the femur displayed medullary bone loss and bone destruction on day 28 after inoculation. Unexpectedly, no significant changes in bone cancer-induced hypersensitivity of pain behaviors were found in P2rx7(-/-) mice, and the changes of pain-related values in CatWalk gait analysis even occurred earlier in P2rx7(-/-) mice, as compared with C57 WT mice. Together with our previous study in rats that blockade of P2X7R significantly alleviated bone cancer pain, it is implied that P2X7R may play different roles in bone cancer pain in different species (e.g. rat vs mouse). These results implicated a huge difference between the pathophysiology discovered in the experimental animal models and that of human disease.


Sujet(s)
Tumeurs osseuses , Douleur cancéreuse , Animaux , Modèles animaux de maladie humaine , Hyperalgésie , Moelle allongée , Souris , Souris de lignée C57BL , Souris knockout , Rats , Rats de lignée LEW , Récepteurs purinergiques P2X7
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...