Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 134(9)2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38530364

RESUMEN

G protein-coupled receptor 37-like 1 (GPR37L1) is an orphan GPCR with largely unknown functions. Here, we report that Gpr37l1/GRP37L1 ranks among the most highly expressed GPCR transcripts in mouse and human dorsal root ganglia (DRGs) and is selectively expressed in satellite glial cells (SGCs). Peripheral neuropathy induced by streptozotoxin (STZ) and paclitaxel (PTX) led to reduced GPR37L1 expression on the plasma membrane in mouse and human DRGs. Transgenic mice with Gpr37l1 deficiency exhibited impaired resolution of neuropathic pain symptoms following PTX- and STZ-induced pain, whereas overexpression of Gpr37l1 in mouse DRGs reversed pain. GPR37L1 is coexpressed with potassium channels, including KCNJ10 (Kir4.1) in mouse SGCs and both KCNJ3 (Kir3.1) and KCNJ10 in human SGCs. GPR37L1 regulates the surface expression and function of the potassium channels. Notably, the proresolving lipid mediator maresin 1 (MaR1) serves as a ligand of GPR37L1 and enhances KCNJ10- or KCNJ3-mediated potassium influx in SGCs through GPR37L1. Chemotherapy suppressed KCNJ10 expression and function in SGCs, which MaR1 rescued through GPR37L1. Finally, genetic analysis revealed that the GPR37L1-E296K variant increased chronic pain risk by destabilizing the protein and impairing the protein's function. Thus, GPR37L1 in SGCs offers a therapeutic target for the protection of neuropathy and chronic pain.


Asunto(s)
Ácidos Docosahexaenoicos , Ganglios Espinales , Neuroglía , Receptores Acoplados a Proteínas G , Transducción de Señal , Animales , Humanos , Masculino , Ratones , Ganglios Espinales/metabolismo , Homeostasis , Ratones Noqueados , Ratones Transgénicos , Neuralgia/metabolismo , Neuralgia/genética , Neuralgia/patología , Neuroglía/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Canales de Potasio de Rectificación Interna/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
2.
bioRxiv ; 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38106084

RESUMEN

G protein coupled receptor 37-like 1 (GPR37L1) is an orphan GPCR and its function remains largely unknown. Here we report that GPR37L1 transcript is highly expressed compared to all known GPCRs in mouse and human dorsal root ganglia (DRGs) and selectively expressed in satellite glial cells (SGCs). Peripheral neuropathy following diabetes and chemotherapy by streptozotocin and paclitaxel resulted in downregulations of surface GPR37L1 in mouse and human DRGs. Transgenic mice with Gpr37l1 deficiency exhibited impaired resolution of neuropathic pain symptom (mechanical allodynia), whereas overexpression of Gpr37l1 in mouse DRGs can reverse neuropathic pain. Notably, GPR37L1 is co-expressed and coupled with potassium channels in SGCs. We found striking species differences in potassium channel expression in SGCs, with predominant expression of KCNJ10 and KCNJ3 in mouse and human SGCs, respectively. GPR37L1 regulates the surface expression and function of KCNJ10 and KCNJ3. We identified the pro-resolving lipid mediator maresin 1 (MaR1) as a GPR37L1 ligand. MaR1 increases KCNJ10/KCNJ3-mediated potassium influx in SGCs via GPR37L1. MaR1 protected chemotherapy-induced suppression of KCNJ13/KCNJ10 expression and function in SGCs. Finally, genetic analysis revealed that the GPR37L1-E296K variant is associated with increased chronic pain risk by destabilizing the protein. Thus, GPR37L1 in SGCs offers a new target for neuropathy protection and pain control.

3.
Neuron ; 111(17): 2709-2726.e9, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37348508

RESUMEN

Programmed death protein 1 (PD-1) and its ligand PD-L1 constitute an immune checkpoint pathway. We report that neuronal PD-1 signaling regulates learning/memory in health and disease. Mice lacking PD-1 (encoded by Pdcd1) exhibit enhanced long-term potentiation (LTP) and memory. Intraventricular administration of anti-mouse PD-1 monoclonal antibody (RMP1-14) potentiated learning and memory. Selective deletion of PD-1 in excitatory neurons (but not microglia) also enhances LTP and memory. Traumatic brain injury (TBI) impairs learning and memory, which is rescued by Pdcd1 deletion or intraventricular PD-1 blockade. Conversely, re-expression of Pdcd1 in PD-1-deficient hippocampal neurons suppresses memory and LTP. Exogenous PD-L1 suppresses learning/memory in mice and the excitability of mouse and NHP hippocampal neurons through PD-1. Notably, neuronal activation suppresses PD-L1 secretion, and PD-L1/PD-1 signaling is distinctly regulated by learning and TBI. Thus, conditions that reduce PD-L1 levels or PD-1 signaling could promote memory in both physiological and pathological conditions.


Asunto(s)
Antígeno B7-H1 , Lesiones Traumáticas del Encéfalo , Humanos , Antígeno B7-H1/metabolismo , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Aprendizaje , Hipocampo/metabolismo , Anticuerpos Monoclonales/metabolismo , Neuronas/metabolismo
4.
Cells ; 12(6)2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36980304

RESUMEN

Preclinical studies have identified glial cells as pivotal players in the genesis and maintenance of neuropathic pain after nerve injury associated with diabetes, chemotherapy, major surgeries, and virus infections. Satellite glial cells (SGCs) in the dorsal root and trigeminal ganglia of the peripheral nervous system (PNS) and astrocytes in the central nervous system (CNS) express similar molecular markers and are protective under physiological conditions. They also serve similar functions in the genesis and maintenance of neuropathic pain, downregulating some of their homeostatic functions and driving pro-inflammatory neuro-glial interactions in the PNS and CNS, i.e., "gliopathy". However, the role of SGCs in neuropathic pain is not simply as "peripheral astrocytes". We delineate how these peripheral and central glia participate in neuropathic pain by producing different mediators, engaging different parts of neurons, and becoming active at different stages following nerve injury. Finally, we highlight the recent findings that SGCs are enriched with proteins related to fatty acid metabolism and signaling such as Apo-E, FABP7, and LPAR1. Targeting SGCs and astrocytes may lead to novel therapeutics for the treatment of neuropathic pain.


Asunto(s)
Astrocitos , Neuralgia , Humanos , Ganglios Espinales/metabolismo , Neuroglía/metabolismo , Neuralgia/metabolismo , Neuronas/metabolismo
5.
Neuron ; 111(4): 450-452, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36796326

RESUMEN

Mechanical distension/stretch in the colon provokes visceral hypersensitivity and pain. In this issue of Neuron, Xie et al. report that mechanosensitive Piezo2 channels, expressed by TRPV1-lineage nociceptors, are involved in visceral mechanical nociception and hypersensitivity.


Asunto(s)
Colon , Canales Iónicos , Dolor , Canales Catiónicos TRPV , Colon/fisiología , Canales Iónicos/fisiología , Neuronas , Nociceptores/fisiología , Canales Catiónicos TRPV/genética
6.
Pain ; 164(6): 1340-1354, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36378290

RESUMEN

ABSTRACT: Specialized proresolving mediators (SPMs) have demonstrated potent analgesic actions in animal models of pathological pain. The actions of SPMs in acute and chronic itch are currently unknown. Recently, n-3 docosapentaenoic acid (DPA) was found to be a substrate for the biosynthesis of several novel families of SPMs and 3-oxa-PD1 n-3 DPA (3-oxa-PD1) is an oxidation-resistant metabolic stable analogue of the n-3 DPA-derived protectin D1 (PD1). In this article, we demonstrate that 3-oxa-PD1 effectively reduces both acute and chronic itch in mouse models. Intrathecal injection of 3-oxa-PD1 (100 ng) reduced acute itch induced by histamine, chloroquine, or morphine. Furthermore, intrathecal 3-oxa-PD1 effectively reduced chronic itch, induced by cutaneous T-cell lymphoma (CTCL), allergic contact dermatitis with dinitrofluorobenzene, and psoriasis by imiquimod. Intratumoral injection of 3-oxa-PD1 also suppressed CTCL-induced chronic itch. Strikingly, the antipruritic effect lasted for several weeks after 1-week intrathecal 3-oxa-PD1 treatment. Whole-cell recordings revealed significant increase in excitatory postsynaptic currents in spinal dorsal horn (SDH) neurons of CTCL mice, but this increase was blocked by 3-oxa-PD1. 3-oxa-PD1 further increased inhibitory postsynaptic currents in SDH neurons of CTCL mice. Cutaneous T-cell lymphoma increased the spinal levels of lipocalin-2 (LCN2), an itch mediator produced by astrocytes. 3-oxa-PD1 suppressed LCN2 production in CTCL mice and LCN2 secretion in astrocytes. Finally, CTCL-induced anxiety was alleviated by intrathecal 3-oxa-PD1. Our findings suggest that 3-oxa-PD1 potently inhibits acute and chronic itch through the regulation of excitatory or inhibitory synaptic transmission and astroglial LCN2 production. Therefore, stable SPM analogs such as 3-oxa-PD1 could be useful to treat pruritus associated with different skin injuries.


Asunto(s)
Astrocitos , Ácidos Grasos Insaturados , Lipocalina 2 , Prurito , Animales , Ratones , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Ácidos Grasos Insaturados/farmacología , Ácidos Grasos Insaturados/uso terapéutico , Lipocalina 2/metabolismo , Linfoma Cutáneo de Células T/complicaciones , Ratones Endogámicos C57BL , Prurito/tratamiento farmacológico , Prurito/etiología , Prurito/metabolismo , Transmisión Sináptica/efectos de los fármacos
7.
Front Immunol ; 12: 787565, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34950149

RESUMEN

The incidence of chronic pain is especially high in women, but the underlying mechanisms remain poorly understood. Interleukin-23 (IL-23) is a pro-inflammatory cytokine and contributes to inflammatory diseases (e.g., arthritis and psoriasis) through dendritic/T cell signaling. Here we examined the IL-23 involvement in sexual dimorphism of pain, using an optogenetic approach in transgenic mice expressing channelrhodopsin-2 (ChR2) in TRPV1-positive nociceptive neurons. In situ hybridization revealed that compared to males, females had a significantly larger portion of small-sized (100-200 µm2) Trpv1+ neurons in dorsal root ganglion (DRG). Blue light stimulation of a hindpaw of transgenic mice induced intensity-dependent spontaneous pain. At the highest intensity, females showed more intense spontaneous pain than males. Intraplantar injection of IL-23 (100 ng) induced mechanical allodynia in females only but had no effects on paw edema. Furthermore, intraplantar IL-23 only potentiated blue light-induced pain in females, and intrathecal injection of IL-23 also potentiated low-dose capsaicin (500 ng) induced spontaneous pain in females but not males. IL-23 expresses in DRG macrophages of both sexes. Intrathecal injection of IL-23 induced significantly greater p38 phosphorylation (p-p38), a marker of nociceptor activation, in DRGs of female mice than male mice. In THP-1 human macrophages estrogen and chemotherapy co-application increased IL-23 secretion, and furthermore, estrogen and IL-23 co-application, but not estrogen and IL-23 alone, significantly increased IL-17A release. These findings suggest a novel role of IL-23 in macrophage signaling and female-dominant pain, including C-fiber-mediated spontaneous pain. Our study has also provided new insight into cytokine-mediated macrophage-nociceptor interactions, in a sex-dependent manner.


Asunto(s)
Ganglios Espinales/efectos de los fármacos , Interleucina-23/toxicidad , Fibras Nerviosas Amielínicas/efectos de los fármacos , Nociceptores/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Dolor/inducido químicamente , Canales Catiónicos TRPV/metabolismo , Animales , Capsaicina , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiopatología , Humanos , Interleucina-17/metabolismo , Luz , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibras Nerviosas Amielínicas/metabolismo , Nociceptores/metabolismo , Optogenética , Dolor/genética , Dolor/metabolismo , Dolor/fisiopatología , Caracteres Sexuales , Células THP-1 , Canales Catiónicos TRPV/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Nat Commun ; 12(1): 4558, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34315904

RESUMEN

Patients with advanced stage cancers frequently suffer from severe pain as a result of bone metastasis and bone destruction, for which there is no efficacious treatment. Here, using multiple mouse models of bone cancer, we report that agonists of the immune regulator STING (stimulator of interferon genes) confer remarkable protection against cancer pain, bone destruction, and local tumor burden. Repeated systemic administration of STING agonists robustly attenuates bone cancer-induced pain and improves locomotor function. Interestingly, STING agonists produce acute pain relief through direct neuronal modulation. Additionally, STING agonists protect against local bone destruction and reduce local tumor burden through modulation of osteoclast and immune cell function in the tumor microenvironment, providing long-term cancer pain relief. Finally, these in vivo effects are dependent on host-intrinsic STING and IFN-I signaling. Overall, STING activation provides unique advantages in controlling bone cancer pain through distinct and synergistic actions on nociceptors, immune cells, and osteoclasts.


Asunto(s)
Neoplasias Óseas/complicaciones , Dolor en Cáncer/etiología , Dolor en Cáncer/inmunología , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Analgésicos/farmacología , Animales , Neoplasias Óseas/sangre , Dolor en Cáncer/sangre , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Fémur/patología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Proteínas de Homeodominio/metabolismo , Hiperalgesia/complicaciones , Interferones/sangre , Interferones/metabolismo , Masculino , Neoplasias Mamarias Animales/complicaciones , Proteínas de la Membrana/agonistas , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Neuronas/efectos de los fármacos , Nocicepción/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteogénesis/efectos de los fármacos , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Xantonas/farmacología
9.
Nat Metab ; 3(6): 735-736, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34140695
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...