Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain Behav Immun ; 116: 203-215, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38070625

RESUMEN

Pain is the most debilitating symptom of knee osteoarthritis (OA) that can even persist after total knee replacement. The severity and duration of pain do not correlate well with joint tissue alterations, suggesting other mechanisms may drive pain persistence in OA. Previous work identified that macrophages accumulate in the dorsal root ganglia (DRG) containing the somas of sensory neurons innervating the injured knee joint in a mouse OA model and acquire a M1-like phenotype to maintain pain. Here we aimed to unravel the mechanisms that govern DRG macrophage accumulation and programming. The accumulation of F4/80+iNOS+ (M1-like) DRG macrophages was detectable at day 3 after mono-iodoacetate (MIA)-induced OA in the mouse. Depletion of macrophages prior to induction of OA resolved pain-like behaviors by day 7 without affecting the initial development of pain-like behaviors. Analysis of DRG transcript identified CXCL11 and myostatin. CXCL11 and myostatin were increased at 3 weeks post OA induction, with CXCL11 expression partially localized in satellite glial cells and myostatin in sensory neurons. Blocking CXCL11 or myostatin prevented the persistence of OA pain, without affecting the initiation of pain. CXCL11 neutralization reduced the number of total and F4/80+iNOS+ DRG macrophages, whilst myostatin inhibition diminished the programming of F4/80+iNOS+ DRG macrophages. Intrathecal injection of recombinant CXCL11 did not induce pain-associated behaviors. In contrast, intrathecal myostatin increased the number of F4/80+iNOS+ DRG macrophages concurrent with the development of mechanical hypersensitivity that was prevented by macrophages depletion or CXCL11 blockade. Finally, myostatin inhibition during established OA, resolved pain and F4/80+iNOS+ macrophage accumulation in the DRG. In conclusion, DRG macrophages maintain OA pain, but are not required for the induction of OA pain. Myostatin is a key ligand in neuro-immune communication that drives the persistence of pain in OA through nervous tissue macrophages and represent a novel therapeutic target for the treatment of OA pain.


Asunto(s)
Tejido Nervioso , Osteoartritis de la Rodilla , Ratas , Ratones , Animales , Miostatina/metabolismo , Ratas Sprague-Dawley , Dolor/metabolismo , Modelos Animales de Enfermedad , Tejido Nervioso/metabolismo , Macrófagos/metabolismo , Ganglios Espinales/metabolismo
2.
PLoS Biol ; 16(2): e2003452, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29444090

RESUMEN

Chronic pain is a debilitating problem, and insights in the neurobiology of chronic pain are needed for the development of novel pain therapies. A genome-wide association study implicated the 5p15.2 region in chronic widespread pain. This region includes the coding region for FAM173B, a functionally uncharacterized protein. We demonstrate here that FAM173B is a mitochondrial lysine methyltransferase that promotes chronic pain. Knockdown and sensory neuron overexpression strategies showed that FAM173B is involved in persistent inflammatory and neuropathic pain via a pathway dependent on its methyltransferase activity. FAM173B methyltransferase activity in sensory neurons hyperpolarized mitochondria and promoted macrophage/microglia activation through a reactive oxygen species-dependent pathway. In summary, we uncover a role for methyltransferase activity of FAM173B in the neurobiology of pain. These results also highlight FAM173B methyltransferase activity as a potential therapeutic target to treat debilitating chronic pain conditions.


Asunto(s)
Dolor Crónico/enzimología , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , Cromosomas Humanos Par 5 , Dolor Crónico/genética , Femenino , Técnicas de Silenciamiento del Gen , Estudio de Asociación del Genoma Completo , Células HEK293 , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Masculino , Ratones Endogámicos C57BL , Microglía/metabolismo , Polimorfismo de Nucleótido Simple , Especies Reactivas de Oxígeno/metabolismo
3.
Cell Mol Life Sci ; 77(23): 4957-4976, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31927610

RESUMEN

Macrophages are key effector cells in obesity-associated inflammation. G protein-coupled receptor kinase 2 (GRK2) is highly expressed in different immune cell types. Using LysM-GRK2+/- mice, we uncover that a reduction of GRK2 levels in myeloid cells prevents the development of glucose intolerance and hyperglycemia after a high fat diet (HFD) through modulation of the macrophage pro-inflammatory profile. Low levels of myeloid GRK2 confer protection against hepatic insulin resistance, steatosis and inflammation. In adipose tissue, pro-inflammatory cytokines are reduced and insulin signaling is preserved. Macrophages from LysM-GRK2+/- mice secrete less pro-inflammatory cytokines when stimulated with lipopolysaccharide (LPS) and their conditioned media has a reduced pathological influence in cultured adipocytes or naïve bone marrow-derived macrophages. Our data indicate that reducing GRK2 levels in myeloid cells, by attenuating pro-inflammatory features of macrophages, has a relevant impact in adipose-liver crosstalk, thus preventing high fat diet-induced metabolic alterations.


Asunto(s)
Tejido Adiposo/metabolismo , Dieta Alta en Grasa , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Hígado/metabolismo , Células Mieloides/metabolismo , Obesidad/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo Blanco/patología , Animales , Medios de Cultivo Condicionados/farmacología , Citoprotección/efectos de los fármacos , Hígado Graso/complicaciones , Hígado Graso/patología , Microbioma Gastrointestinal/efectos de los fármacos , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Hipertrofia , Inflamación/patología , Insulina/metabolismo , Resistencia a la Insulina , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Células Mieloides/efectos de los fármacos , Obesidad/complicaciones , Transducción de Señal/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
4.
J Biol Chem ; 294(31): 11654-11664, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31213526

RESUMEN

Lysine methylation is a common posttranslational modification of nuclear and cytoplasmic proteins but is also present in mitochondria. The human protein denoted "family with sequence similarity 173 member B" (FAM173B) was recently uncovered as a mitochondrial lysine (K)-specific methyltransferase (KMT) targeting the c-subunit of mitochondrial ATP synthase (ATPSc), and was therefore renamed ATPSc-KMT. We here set out to investigate the biochemical function of its yet uncharacterized paralogue FAM173A. We demonstrate that FAM173A localizes to mitochondria, mediated by a noncanonical targeting sequence that is partially retained in the mature protein. Immunoblotting analysis using methyllysine-specific antibodies revealed that FAM173A knock-out (KO) abrogates lysine methylation of a single mitochondrial protein in human cells. Mass spectrometry analysis identified this protein as adenine nucleotide translocase (ANT), represented by two highly similar isoforms ANT2 and ANT3. We found that methylation occurs at Lys-52 of ANT, which was previously reported to be trimethylated. Complementation of KO cells with WT or enzyme-dead FAM173A indicated that the enzymatic activity of FAM173A is required for ANT methylation at Lys-52 to occur. Both in human cells and in rat organs, Lys-52 was exclusively trimethylated, indicating that this modification is constitutive, rather than regulatory and dynamic. Moreover, FAM173A-deficient cells displayed increased mitochondrial respiration compared with FAM173A-proficient cells. In summary, we demonstrate that FAM173A is the long-sought KMT responsible for ANT methylation at Lys-52, and point out the functional significance of Lys-52 methylation in ANT. Based on the established naming nomenclature for KMTs, we propose to rename FAM173A to ANT-KMT (gene name ANTKMT).


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Mitocondrias/metabolismo , Translocasas Mitocondriales de ADP y ATP/metabolismo , Proteínas Mitocondriales/metabolismo , Proteína Metiltransferasas/metabolismo , Secuencia de Aminoácidos , Animales , Cromatografía Líquida de Alta Presión , Células HeLa , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Hígado/metabolismo , Lisina/metabolismo , Espectrometría de Masas , Metilación , Mitocondrias/enzimología , Proteínas Mitocondriales/genética , Péptidos/análisis , Proteína Metiltransferasas/genética , Ratas , Alineación de Secuencia
5.
J Biol Chem ; 294(4): 1128-1141, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30530489

RESUMEN

Lysine methylation is an important post-translational modification that is also present on mitochondrial proteins, but the mitochondrial lysine-specific methyltransferases (KMTs) responsible for modification are in most cases unknown. Here, we set out to determine the function of human family with sequence similarity 173 member B (FAM173B), a mitochondrial methyltransferase (MTase) reported to promote chronic pain. Using bioinformatics analyses and biochemical assays, we found that FAM173B contains an atypical, noncleavable mitochondrial targeting sequence responsible for its localization to mitochondria. Interestingly, CRISPR/Cas9-mediated KO of FAM173B in mammalian cells abrogated trimethylation of Lys-43 in ATP synthase c-subunit (ATPSc), a modification previously reported as ubiquitous among metazoans. ATPSc methylation was restored by complementing the KO cells with enzymatically active human FAM173B or with a putative FAM173B orthologue from the nematode Caenorhabditis elegans Interestingly, lack of Lys-43 methylation caused aberrant incorporation of ATPSc into the ATP synthase complex and resulted in decreased ATP-generating ability of the complex, as well as decreased mitochondrial respiration. In summary, we have identified FAM173B as the long-sought KMT responsible for methylation of ATPSc, a key protein in cellular ATP production, and have demonstrated functional significance of ATPSc methylation. We suggest renaming FAM173B to ATPSc-KMT (gene name ATPSCKMT).


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Lisina/metabolismo , Mitocondrias/enzimología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Animales , Línea Celular , Biología Computacional , Células HeLa , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Metilación , Ratones , Mitocondrias/metabolismo
6.
Rheumatology (Oxford) ; 57(3): 429-440, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28968842

RESUMEN

Chronic pain is a major debilitating condition that is difficult to treat. Although chronic pain may appear to be a disorder of the nervous system, crucial roles for immune cells and their mediators have been identified as important contributors in various types of pain. This review focuses on how the immune system regulates pain and discusses the emerging roles of immune cells in the initiation or maintenance of chronic pain. We highlight which immune cells infiltrate damaged nerves, the dorsal root ganglia, spinal cord and tissues around free nerve endings and discuss through which mechanisms they control pain. Finally we discuss emerging roles of the immune system in resolving pain and how the immune system contributes to the transition from acute to chronic pain. We propose that targeting some of these immune processes may provide novel therapeutic opportunities for the treatment of chronic pain.


Asunto(s)
Dolor Agudo/inmunología , Dolor Crónico/inmunología , Sistema Inmunológico/fisiología , Inmunidad Celular , Ganglios Espinales/inmunología , Humanos , Médula Espinal/inmunología
7.
J Neurosci ; 36(28): 7353-63, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27413147

RESUMEN

UNLABELLED: Chronic pain is a major clinical problem that is difficult to treat and requires novel therapies. Although most pain therapies primarily target neurons, neuroinflammatory processes characterized by spinal cord and dorsal root ganglion production of proinflammatory cytokines play an important role in persistent pain states and represent potential therapeutic targets. Anti-inflammatory cytokines are attractive candidates to regulate aberrant neuroinflammatory processes, but the therapeutic potential of these cytokines as stand-alone drugs is limited. Their optimal function requires concerted actions with other regulatory cytokines, and their relatively small size causes rapid clearance. To overcome these limitations, we developed a fusion protein of the anti-inflammatory cytokines interleukin 4 (IL4) and IL10. The IL4-10 fusion protein is a 70 kDa glycosylated dimeric protein that retains the functional activity of both cytokine moieties. Intrathecal administration of IL4-10 dose-dependently inhibited persistent inflammatory pain in mice: three IL4-10 injections induced full resolution of inflammatory pain in two different mouse models of persistent inflammatory pain. Both cytokine moieties were required for optimal effects. The IL4-10 fusion protein was more effective than the individual cytokines or IL4 plus IL10 combination therapy and also inhibited allodynia in a mouse model of neuropathic pain. Mechanistically, IL4-10 inhibited the activity of glial cells and reduced spinal cord and dorsal root ganglion cytokine levels without affecting paw inflammation. In conclusion, we developed a novel fusion protein with improved efficacy to treat pain, compared with wild-type anti-inflammatory cytokines. The IL4-10 fusion protein has potential as a treatment for persistent inflammatory pain. SIGNIFICANCE STATEMENT: The treatment of chronic pain is a major clinical and societal challenge. Current therapies to treat persistent pain states are limited and often cause major side effects. Therefore, novel analgesic treatments are urgently needed. In search of a novel drug to treat chronic pain, we developed a fusion protein consisting of two prototypic regulatory cytokines, interleukin 4 (IL4) and IL10. The work presented in this manuscript shows that this IL4-10 fusion protein overcomes some major therapeutic limitations of pain treatment with individual cytokines. The IL4-10 fusion protein induces full resolution of persistent inflammatory pain in two different mouse models. These novel findings are significant, as they highlight the IL4-10 fusion protein as a long-needed potential new drug to stop persistent pain states.


Asunto(s)
Analgésicos/uso terapéutico , Inflamación/complicaciones , Interleucina-10/uso terapéutico , Interleucina-4/uso terapéutico , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Animales , Carragenina/toxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Adyuvante de Freund/toxicidad , Humanos , Inflamación/inducido químicamente , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Manejo del Dolor , Umbral del Dolor/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Médula Espinal/citología , Resultado del Tratamiento
8.
Biochem J ; 459(3): 427-39, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24517375

RESUMEN

The MAPK (mitogen-activated protein kinase) p38 is an important mediator of inflammation and of inflammatory and neuropathic pain. We have described recently that docking-groove-dependent interactions are important for p38 MAPK-mediated signal transduction. Thus virtual screening was performed to identify putative docking-groove-targeted p38 MAPK inhibitors. Several compounds of the benzo-oxadiazol family were identified with low micromolar inhibitory activity both in a p38 MAPK activity assay, and in THP-1 human monocytes acting as inhibitors of LPS (lipopolysaccharide)-induced TNFα (tumour necrosis factor α) secretion. Positions 2 and 5 in the phenyl ring are essential for the described inhibitory activity with a chloride in position 5 and a methyl group in position 2 yielding the best results, giving an IC50 value of 1.8 µM (FGA-19 compound). Notably, FGA-19 exerted a potent and long-lasting analgesic effect in vivo when tested in a mouse model of inflammatory hyperalgesia. A single intrathecal injection of FGA-19 completely resolved hyperalgesia, being 10-fold as potent and displaying longer lasting effects than the established p38 MAPK inhibitor SB239063. FGA-19 also reversed persistent pain in a model of post-inflammatory hyperalgesia in LysM (lysozyme M)-GRK2 (G-protein-coupled-receptor kinase)(+/-) mice. These potent in vivo effects suggested p38 MAPK docking-site-targeted inhibitors as a potential novel strategy for the treatment of inflammatory pain.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Hiperalgesia/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Oxadiazoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Analgésicos/química , Analgésicos/metabolismo , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/uso terapéutico , Línea Celular , Células Cultivadas , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Hiperalgesia/inmunología , Hiperalgesia/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Monocitos/inmunología , Monocitos/metabolismo , Oxadiazoles/química , Oxadiazoles/metabolismo , Oxadiazoles/uso terapéutico , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Distribución Aleatoria , Relación Estructura-Actividad , Proteínas Quinasas p38 Activadas por Mitógenos/química , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Stroke ; 44(5): 1426-32, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23539530

RESUMEN

BACKGROUND AND PURPOSE: Brain injury caused by stroke is a frequent cause of perinatal morbidity and mortality with limited therapeutic options. Mesenchymal stem cells (MSC) have been shown to improve outcome after neonatal hypoxic-ischemic brain injury mainly by secretion of growth factors stimulating repair processes. We investigated whether MSC treatment improves recovery after neonatal stroke and whether MSC overexpressing brain-derived neurotrophic factor (MSC-BDNF) further enhances recovery. METHODS: We performed 1.5-hour transient middle cerebral artery occlusion in 10-day-old rats. Three days after reperfusion, pups with evidence of injury by diffusion-weighted MRI were treated intranasally with MSC, MSC-BDNF, or vehicle. To determine the effect of MSC treatment, brain damage, sensorimotor function, and cerebral cell proliferation were analyzed. RESULTS: Intranasal delivery of MSC- and MSC-BDNF significantly reduced infarct size and gray matter loss in comparison with vehicle-treated rats without any significant difference between MSC- and MSC-BDNF-treatment. Treatment with MSC-BDNF significantly reduced white matter loss with no significant difference between MSC- and MSC-BDNF-treatment. Motor deficits were also improved by MSC treatment when compared with vehicle-treated rats. MSC-BDNF-treatment resulted in an additional significant improvement of motor deficits 14 days after middle cerebral artery occlusion, but there was no significant difference between MSC or MSC-BDNF 28 days after middle cerebral artery occlusion. Furthermore, treatment with either MSC or MSC-BDNF induced long-lasting cell proliferation in the ischemic hemisphere. CONCLUSIONS: Intranasal administration of MSC after neonatal stroke is a promising therapy for treatment of neonatal stroke. In this experimental paradigm, MSC- and BNDF-hypersecreting MSC are equally effective in reducing ischemic brain damage.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Encéfalo/patología , Infarto de la Arteria Cerebral Media/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Accidente Cerebrovascular/terapia , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Fibras Nerviosas Mielínicas/patología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/patología
10.
Neurobiol Dis ; 54: 206-15, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23313319

RESUMEN

G protein-coupled receptor (GPCR) kinase 2 (GRK2) regulates cellular signaling via desensitization of GPCRs and by direct interaction with intracellular signaling molecules. We recently described that ischemic brain injury decreases cerebral GRK2 levels. Here we studied the effect of astrocyte GRK2-deficiency on neonatal brain damage in vivo. As astrocytes protect neurons by taking up glutamate via plasma-membrane transporters, we also studied the effect of GRK2 on the localization of the GLutamate ASpartate Transporter (GLAST). Brain damage induced by hypoxia-ischemia was significantly reduced in GFAP-GRK2(+/-) mice, which have a 60% reduction in astrocyte GRK2 compared to GFAP-WT littermates. In addition, GRK2-deficient astrocytes have higher plasma-membrane levels of GLAST and an increased capacity to take up glutamate in vitro. In search for the mechanism by which GRK2 regulates GLAST expression, we observed increased GFAP levels in GRK2-deficient astrocytes. GFAP and the cytoskeletal protein ezrin are known regulators of GLAST localization. In line with this evidence, GRK2-deficiency reduced phosphorylation of the GRK2 substrate ezrin and enforced plasma-membrane GLAST association after stimulation with the group I mGluR-agonist DHPG. When ezrin was silenced, the enhanced plasma-membrane GLAST association in DHPG-exposed GRK2-deficient astrocytes was prevented. In conclusion, we identified a novel role of astrocyte GRK2 in regulating plasma-membrane GLAST localization via an ezrin-dependent route. We demonstrate that the 60% reduction in astrocyte GRK2 protein level that is observed in GFAP-GRK2(+/-) mice is sufficient to significantly reduce neonatal ischemic brain damage. These findings underline the critical role of GRK2 regulation in astrocytes for dampening the extent of brain damage after ischemia.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Astrocitos/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Ácido Glutámico/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Proteínas del Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Interferente Pequeño
11.
Ann Rheum Dis ; 72(3): 427-36, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22956598

RESUMEN

BACKGROUND AND OBJECTIVES: Chronic widespread pain (CWP) is a common disorder affecting ∼10% of the general population and has an estimated heritability of 48-52%. In the first large-scale genome-wide association study (GWAS) meta-analysis, we aimed to identify common genetic variants associated with CWP. METHODS: We conducted a GWAS meta-analysis in 1308 female CWP cases and 5791 controls of European descent, and replicated the effects of the genetic variants with suggestive evidence for association in 1480 CWP cases and 7989 controls. Subsequently, we studied gene expression levels of the nearest genes in two chronic inflammatory pain mouse models, and examined 92 genetic variants previously described associated with pain. RESULTS: The minor C-allele of rs13361160 on chromosome 5p15.2, located upstream of chaperonin-containing-TCP1-complex-5 gene (CCT5) and downstream of FAM173B, was found to be associated with a 30% higher risk of CWP (minor allele frequency=43%; OR=1.30, 95% CI 1.19 to 1.42, p=1.2×10(-8)). Combined with the replication, we observed a slightly attenuated OR of 1.17 (95% CI 1.10 to 1.24, p=4.7×10(-7)) with moderate heterogeneity (I2=28.4%). However, in a sensitivity analysis that only allowed studies with joint-specific pain, the combined association was genome-wide significant (OR=1.23, 95% CI 1.14 to 1.32, p=3.4×10(-8), I2=0%). Expression levels of Cct5 and Fam173b in mice with inflammatory pain were higher in the lumbar spinal cord, not in the lumbar dorsal root ganglions, compared to mice without pain. None of the 92 genetic variants previously described were significantly associated with pain (p>7.7×10(-4)). CONCLUSIONS: We identified a common genetic variant on chromosome 5p15.2 associated with joint-specific CWP in humans. This work suggests that CCT5 and FAM173B are promising targets in the regulation of pain.


Asunto(s)
Cromosomas Humanos Par 5/genética , Dolor Crónico/genética , Estudio de Asociación del Genoma Completo , Animales , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Ratones , Polimorfismo de Nucleótido Simple
12.
Immunother Adv ; 3(1): ltad022, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38047118

RESUMEN

Pain is one of the most debilitating symptoms in rheumatic diseases. Pain often persists after total knee replacement in osteoarthritis, or when inflammation is minimal/absent in rheumatoid arthritis. This suggests that pain transitions to a chronic state independent of the original damage/inflammation. Mitochondrial dysfunction in the nervous system promotes chronic pain and is linked to NLRP3 inflammasome activation. Therefore, we investigated the role of mitochondrial dysfunction and NLRP3 inflammasome activation in the transition from acute to persistent inflammation-induced nociplastic pain and in persistent monoiodoacetate-induced osteoarthritis pain. Intraplantar injection of carrageenan in mice induced transient inflammatory pain that resolved within 7 days. A subsequent intraplantar PGE2 injection induced persistent mechanical hypersensitivity, while in naive mice it resolved within one day. Thus, this initial transient inflammation induced maladaptive nociceptor neuroplasticity, so-called hyperalgesic priming. At Day 7, when mice were primed, expression of NLRP3 inflammasome pathway components was increased, and dorsal root ganglia (DRG) neurons displayed signs of activated NLRP3 inflammasome. Inhibition of NLRP3 inflammasome with MCC950 prevented the transition from acute to chronic pain in this hyperalgesic priming model. In mice with persistent monoiodoacetate-induced osteoarthritis pain, DRG neurons displayed signs of mitochondrial oxidative stress and NLRP3 inflammasome activation. Blocking NLRP3 inflammasome activity attenuated established osteoarthritis pain. In males, NLPR3 inhibition had longer-lasting effects than in females. Overall, these data suggest that NLRP3 inflammasome activation in sensory neurons, potentially caused by neuronal oxidative stress, promotes development of persistent inflammatory and osteoarthritis pain. Therefore, targeting NLRP3 inflammasome pathway may be a promising approach to treat chronic pain.

13.
Cell Rep Med ; 4(11): 101265, 2023 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-37944527

RESUMEN

Pain often persists in patients with an inflammatory disease, even when inflammation has subsided. The molecular mechanisms leading to this failure in pain resolution and the transition to chronic pain are poorly understood. Mitochondrial dysfunction in sensory neurons links to chronic pain, but its role in resolution of inflammatory pain is unclear. Transient inflammation causes neuronal plasticity, called hyperalgesic priming, which impairs resolution of pain induced by a subsequent inflammatory stimulus. We identify that hyperalgesic priming in mice increases the expression of a mitochondrial protein (ATPSc-KMT) and causes mitochondrial and metabolic disturbances in sensory neurons. Inhibition of mitochondrial respiration, knockdown of ATPSCKMT expression, or supplementation of the affected metabolite is sufficient to restore resolution of inflammatory pain and prevents chronic pain development. Thus, inflammation-induced mitochondrial-dependent disturbances in sensory neurons predispose to a failure in resolution of inflammatory pain and development of chronic pain.


Asunto(s)
Dolor Crónico , Humanos , Ratones , Animales , Dolor Crónico/inducido químicamente , Dolor Crónico/metabolismo , Células Receptoras Sensoriales/metabolismo , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Inflamación/metabolismo , Mitocondrias/metabolismo
14.
Mol Med ; 18: 556-64, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22331028

RESUMEN

The molecular mechanisms determining magnitude and duration of inflammatory pain are still unclear. We assessed the contribution of G protein-coupled receptor kinase (GRK)-6 to inflammatory hyperalgesia in mice. We showed that GRK6 is a critical regulator of severity and duration of cytokine-induced hyperalgesia. In GRK6⁻/⁻ mice, a significantly lower dose (100 times lower) of intraplantar interleukin (IL)-1ß was sufficient to induce hyperalgesia compared with wild-type (WT) mice. In addition, IL-1ß hyperalgesia lasted much longer in GRK6⁻/⁻ mice than in WT mice (8 d in GRK6⁻/⁻ versus 6 h in WT mice). Tumor necrosis factor (TNF)-α-induced hyperalgesia was also enhanced and prolonged in GRK6⁻/⁻ mice. In vitro, IL-1ß-induced p38 phosphorylation in GRK6⁻/⁻ dorsal root ganglion (DRG) neurons was increased compared with WT neurons. In contrast, IL-1ß only induced activation of the phosphatidylinositol (PI) 3-kinase/Akt pathway in WT neurons, but not in GRK6⁻/⁻ neurons. In vivo, p38 inhibition attenuated IL-1ß- and TNF-α-induced hyperalgesia in both genotypes. Notably, however, whereas PI 3-kinase inhibition enhanced and prolonged hyperalgesia in WT mice, it did not have any effect in GRK6-deficient mice. The capacity of GRK6 to regulate pain responses was also apparent in carrageenan-induced hyperalgesia, since thermal and mechanical hypersensitivity was significantly prolonged in GRK6⁻/⁻ mice. Finally, GRK6 expression was reduced in DRGs of mice with chronic neuropathic or inflammatory pain. Collectively, these findings underline the potential role of GRK6 in pathological pain. We propose the novel concept that GRK6 acts as a kinase that constrains neuronal responsiveness to IL-1ß and TNF-α and cytokine-induced hyperalgesia via biased cytokine-induced p38 and PI 3-kinase/Akt activation.


Asunto(s)
Citocinas/toxicidad , Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Hiperalgesia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Citocinas/administración & dosificación , Dinoprostona/administración & dosificación , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Femenino , Quinasas de Receptores Acoplados a Proteína-G/genética , Regulación de la Expresión Génica , Hiperalgesia/inducido químicamente , Hiperalgesia/genética , Inflamación/genética , Inflamación/metabolismo , Interleucina-1beta/administración & dosificación , Ratones , Ratones Noqueados , Neuralgia/genética , Neuralgia/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/administración & dosificación , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
15.
J Neuroinflammation ; 9: 143, 2012 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-22731384

RESUMEN

BACKGROUND: Chronic pain is often associated with microglia activation in the spinal cord. We recently showed that microglial levels of the kinase G protein-coupled receptor kinase (GRK)2 are reduced in models of chronic pain. We also found that mice with a cell-specific reduction of around 50% in GRK2 level in microglia/macrophages (LysM-GRK2+/- mice) develop prolonged inflammatory hyperalgesia concomitantly with ongoing spinal microglia/macrophage activation. The microRNA miR-124 is thought to keep microglia/macrophages in brain and spinal cord in a quiescent state. In the present study, we investigated the contribution of miR-124 to regulation of hyperalgesia and microglia/macrophage activation in GRK2-deficient mice. In addition, we investigated the effect of miR-124 on chronic inflammatory and neuropathic pain in wild-type (WT) mice. METHODS: Hyperalgesia was induced by intraplantar IL-1ß in WT and LysM-GRK2+/- mice. We determined spinal cord microglia/macrophage miR-124 expression and levels of pro-inflammatory M1 and anti-inflammatory M2 activation markers. The effect of intrathecal miR-124 treatment on IL-1ß-induced hyperalgesia and spinal M1/M2 phenotype, and on carrageenan-induced and spared nerve injury-induced chronic hyperalgesia in WT mice was analyzed. RESULTS: Transition from acute to persistent hyperalgesia in LysM-GRK2+/- mice is associated with reduced spinal cord microglia miR-124 levels. In our LysM-GRK2+/- mice, there was a switch towards a pro-inflammatory M1 phenotype together with increased pro-inflammatory cytokine production. Intrathecal administration of miR-124 completely prevented the transition to persistent pain in response to IL-1ß in LysM-GRK2+/- mice. The miR-124 treatment also normalized expression of spinal M1/M2 markers of LysM-GRK2+/- mice. Moreover, intrathecal miR-124 treatment reversed the persistent hyperalgesia induced by carrageenan in WT mice and prevented development of mechanical allodynia in the spared nerve injury model of chronic neuropathic pain in WT mice. CONCLUSIONS: We present the first evidence that intrathecal miR-124 treatment can be used to prevent and treat persistent inflammatory and neuropathic pain. In addition, we show for the first time that persistent hyperalgesia in GRK2-deficient mice is associated with an increased ratio of M1/M2 type markers in spinal cord microglia/macrophages, which is restored by miR-124 treatment. We propose that intrathecal miR-124 treatment might be a powerful novel treatment for pathological chronic pain with persistent microglia activation.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/deficiencia , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , MicroARNs/uso terapéutico , Animales , Femenino , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Hiperalgesia/genética , Inyecciones Espinales , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/administración & dosificación , MicroARNs/genética , Microglía/efectos de los fármacos , Microglía/metabolismo , Resultado del Tratamiento
16.
Front Pain Res (Lausanne) ; 3: 1013577, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36324872

RESUMEN

Rheumatic diseases, such as osteoarthritis and rheumatoid arthritis, affect over 750 million people worldwide and contribute to approximately 40% of chronic pain cases. Inflammation and tissue damage contribute to pain in rheumatic diseases, but pain often persists even when inflammation/damage is resolved. Mechanisms that cause this persistent pain are still unclear. Mitochondria are essential for a myriad of cellular processes and regulate neuronal functions. Mitochondrial dysfunction has been implicated in multiple neurological disorders, but its role in sensory processing and pain in rheumatic diseases is relatively unexplored. This review provides a comprehensive understanding of how mitochondrial dysfunction connects inflammation and damage-associated pathways to neuronal sensitization and persistent pain. To provide an overall framework on how mitochondria control pain, we explored recent evidence in inflammatory and neuropathic pain conditions. Mitochondria have intrinsic quality control mechanisms to prevent functional deficits and cellular damage. We will discuss the link between neuronal activity, mitochondrial dysfunction and chronic pain. Lastly, pharmacological strategies aimed at reestablishing mitochondrial functions or boosting mitochondrial dynamics as therapeutic interventions for chronic pain are discussed. The evidence presented in this review shows that mitochondria dysfunction may play a role in rheumatic pain. The dysfunction is not restricted to neuronal cells in the peripheral and central nervous system, but also includes blood cells and cells at the joint level that may affect pain pathways indirectly. Pre-clinical and clinical data suggest that modulation of mitochondrial functions can be used to attenuate or eliminate pain, which could be beneficial for multiple rheumatic diseases.

17.
Neuron ; 110(4): 613-626.e9, 2022 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-34921782

RESUMEN

The current paradigm is that inflammatory pain passively resolves following the cessation of inflammation. Yet, in a substantial proportion of patients with inflammatory diseases, resolution of inflammation is not sufficient to resolve pain, resulting in chronic pain. Mechanistic insight into how inflammatory pain is resolved is lacking. Here, we show that macrophages actively control resolution of inflammatory pain remotely from the site of inflammation by transferring mitochondria to sensory neurons. During resolution of inflammatory pain in mice, M2-like macrophages infiltrate the dorsal root ganglia that contain the somata of sensory neurons, concurrent with the recovery of oxidative phosphorylation in sensory neurons. The resolution of pain and the transfer of mitochondria requires expression of CD200 receptor (CD200R) on macrophages and the non-canonical CD200R-ligand iSec1 on sensory neurons. Our data reveal a novel mechanism for active resolution of inflammatory pain.


Asunto(s)
Macrófagos , Células Receptoras Sensoriales , Animales , Ganglios Espinales/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Mitocondrias , Dolor/metabolismo , Células Receptoras Sensoriales/metabolismo
18.
J Neurosci ; 30(38): 12806-15, 2010 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-20861385

RESUMEN

Hyperexcitability of peripheral nociceptive pathways is often associated with inflammation and is an important mechanism underlying inflammatory pain. Here we describe a completely novel mechanism via which nociceptor G-protein-coupled receptor kinase 2 (GRK2) contributes to regulation of inflammatory hyperalgesia. We show that nociceptor GRK2 is downregulated during inflammation. In addition, we show for the first time that prostaglandin E2 (PGE2)-induced hyperalgesia is prolonged from <6 h in wild-type (WT) mice to 3 d in mice with low GRK2 in Nav1.8+ nociceptors (SNS-GRK2+/- mice). This prolongation of PGE2 hyperalgesia in SNS-GRK2+/- mice does not depend on changes in the sensitivity of the prostaglandin receptors because prolonged hyperalgesia also developed in response to 8-Br-cAMP. PGE2 or cAMP-induced hyperalgesia in WT mice is PKA dependent. However, PKA activity is not required for hyperalgesia in SNS-GRK2+/- mice. SNS-GRK2+/- mice developed prolonged hyperalgesia in response to the Exchange proteins directly activated by cAMP (Epac) activator 8-pCPT-2'-O-Me-cAMP (8-pCPT). Coimmunoprecipitation experiments showed that GRK2 binds to Epac1. In vitro, GRK2 deficiency increased 8-pCPT-induced activation of the downstream effector of Epac, Rap1, and extracellular signal-regulated kinase (ERK). In vivo, inhibition of MEK1 or PKCε prevented prolonged PGE2, 8-Br-cAMP, and 8-pCPT hyperalgesia in SNS-GRK2+/- mice. In conclusion, we discovered GRK2 as a novel Epac1-interacting protein. A reduction in the cellular level of GRK2 enhances activation of the Epac-Rap1 pathway. In vivo, low nociceptor GRK2 leads to prolonged inflammatory hyperalgesia via biased cAMP signaling from PKA to Epac-Rap1, ERK/PKCε pathways.


Asunto(s)
AMP Cíclico/metabolismo , Dinoprostona/farmacología , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hiperalgesia/metabolismo , Inflamación/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Western Blotting , Línea Celular , Células Cultivadas , Dinoprostona/metabolismo , Regulación hacia Abajo , Femenino , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Humanos , Hiperalgesia/inducido químicamente , Inmunoprecipitación , Inflamación/inducido químicamente , Ratones , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal/fisiología
19.
J Neurosci ; 30(6): 2138-49, 2010 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-20147541

RESUMEN

Chronic pain associated with inflammation is a common clinical problem, and the underlying mechanisms have only begun to be unraveled. GRK2 regulates cellular signaling by promoting G-protein-coupled receptor (GPCR) desensitization and direct interaction with downstream kinases including p38. The aim of this study was to determine the contribution of GRK2 to regulation of inflammatory pain and to unravel the underlying mechanism. GRK2(+/-) mice with an approximately 50% reduction in GRK2 developed increased and markedly prolonged thermal hyperalgesia and mechanical allodynia after carrageenan-induced paw inflammation or after intraplantar injection of the GPCR-binding chemokine CCL3. The effect of reduced GRK2 in specific cells was investigated using Cre-Lox technology. Carrageenan- or CCL3-induced hyperalgesia was increased but not prolonged in mice with decreased GRK2 only in Na(v)1.8 nociceptors. In vitro, reduced neuronal GRK2 enhanced CCL3-induced TRPV1 sensitization. In vivo, CCL3-induced acute hyperalgesia in GRK2(+/-) mice was mediated via TRPV1. Reduced GRK2 in microglia/monocytes only was required and sufficient to transform acute carrageenan- or CCL3-induced hyperalgesia into chronic hyperalgesia. Chronic hyperalgesia in GRK2(+/-) mice was associated with ongoing microglial activation and increased phospho-p38 and tumor necrosis factor alpha (TNF-alpha) in the spinal cord. Inhibition of spinal cord microglial, p38, or TNF-alpha activity by intrathecal administration of specific inhibitors reversed ongoing hyperalgesia in GRK2(+/-) mice. Microglia/macrophage GRK2 expression was reduced in the lumbar ipsilateral spinal cord during neuropathic pain, underlining the pathophysiological relevance of microglial GRK2. Thus, we identified completely novel cell-specific roles of GRK2 in regulating acute and chronic inflammatory hyperalgesia.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Dolor/enzimología , Dolor/fisiopatología , Animales , Astrocitos/metabolismo , Células Cultivadas , Quimiocina CCL3/farmacología , Quimiocina CCL3/fisiología , Femenino , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Hiperalgesia/enzimología , Hiperalgesia/fisiopatología , Inflamación/enzimología , Inflamación/fisiopatología , Macrófagos/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/enzimología , Dolor/inmunología , Enfermedades del Sistema Nervioso Periférico/enzimología , Enfermedades del Sistema Nervioso Periférico/inmunología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/enzimología , Médula Espinal/enzimología , Canales Catiónicos TRPV/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
20.
Brain Behav Immun ; 25(6): 1055-60, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21473908

RESUMEN

Pain is a hallmark of tissue damage and inflammation promoting tissue protection and thereby contributing to repair. Therefore, transient acute pain is an important feature of the adaptive response to damage. However, in a significant number of cases, pain persists for months to years after the problem that originally caused the pain has resolved. Such chronic pain is maladaptive as it no longer serves a protective aim. Chronic pain is debilitating, both physiologically and psychologically, and treatments to provide relief from chronic pain are often ineffective. The neurobiological mechanisms underlying the transition from adaptive acute pain to maladaptive chronic pain are only partially understood. In this review, we will summarize recent evidence that a kinase known as G protein-coupled receptor kinase (GRK2) is a key regulator of the transition from acute to chronic inflammatory pain. Our recent studies have shown that mice with a reduction in the cellular level of GRK2 develop chronic hyperalgesia in response to inflammatory mediators that induce only transient hyperalgesia in WT mice. This finding is clinically relevant because rodent models of chronic pain are associated with reduced cellular levels of GRK2. We propose that GRK2 is a newly discovered major player in the regulation of chronic pain. The pathways regulated by this kinase may open up new avenues for development of treatment strategies that target the cause, and not the symptoms of chronic pain.


Asunto(s)
Dolor Crónico/enzimología , Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Inflamación/fisiopatología , Microglía/enzimología , Neuralgia/enzimología , Dolor Agudo/enzimología , Dolor Agudo/fisiopatología , Animales , Dolor Crónico/fisiopatología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Predicción , Quinasa 2 del Receptor Acoplado a Proteína-G/deficiencia , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Regulación Enzimológica de la Expresión Génica , Humanos , Hiperalgesia/enzimología , Hiperalgesia/fisiopatología , Macrófagos/enzimología , Ratones , Ratones Noqueados , Ratones Transgénicos , Microglía/metabolismo , Neuralgia/fisiopatología , Receptores de Interleucina-1/fisiología , Nervio Ciático/lesiones , Transducción de Señal , Médula Espinal/patología , Médula Espinal/fisiopatología , Nervios Espinales/lesiones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA