Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 80(6): 940-954.e6, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33202251

RESUMEN

Mechanisms that control mobilization of cytosolic calcium [Ca2+]i are key for regulation of numerous eukaryotic cell functions. One such paradigmatic mechanism involves activation of phospholipase Cß (PLCß) enzymes by G protein ßγ subunits from activated Gαi-Gßγ heterotrimers. Here, we report identification of a master switch to enable this control for PLCß enzymes in living cells. We find that the Gαi-Gßγ-PLCß-Ca2+ signaling module is entirely dependent on the presence of active Gαq. If Gαq is pharmacologically inhibited or genetically ablated, Gßγ can bind to PLCß but does not elicit Ca2+ signals. Removal of an auto-inhibitory linker that occludes the active site of the enzyme is required and sufficient to empower "stand-alone control" of PLCß by Gßγ. This dependence of Gi-Gßγ-Ca2+ on Gαq places an entire signaling branch of G-protein-coupled receptors (GPCRs) under hierarchical control of Gq and changes our understanding of how Gi-GPCRs trigger [Ca2+]i via PLCß enzymes.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/genética , Proteínas de Unión al GTP Heterotriméricas/genética , Fosfolipasa C beta/genética , Calcio/metabolismo , Señalización del Calcio/genética , Citosol/metabolismo , Células HEK293 , Humanos , Unión Proteica/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/genética
2.
Glia ; 71(11): 2499-2510, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37278537

RESUMEN

In the central nervous system, oligodendrocytes (OLs) produce myelin sheaths that provide trophic support to neuronal axons and increase the propagation speed of action potential. OLs are constantly generated from OL precursor cells (OPCs) throughout life span. The production of myelinating OLs consists of three canonical stages: OPCs, newly-formed OLs (NFOs), and mature myelinating OLs. Recently, single-cell RNA transcriptomic analyses identified a new population of oligodendroglial cells, namely differentiation committed OPCs (COPs). COPs represent a critical intermediate population between OPCs and NFOs, as revealed by specific expression of G-protein coupled receptor 17 (GPR17). The dysregulation of COPs leads to the remyelination failure in demyelinating diseases and impairs the replacement of lost myelin sheaths due to aging. Hence, understanding the development of COPs and their underlying regulatory network will be helpful in establishing new strategies for promoting myelin repair in demyelinating diseases. This review summarizes the current knowledge on the development and functions of COPs under both physiological and pathological conditions. Overall, COPs function as "checkpoints" to prevent inappropriate precocious OL differentiation and myelination through expressing distinct regulatory factors. Deepening our understanding of COPs may not only advance our knowledge of how OL lineage progresses during development, but also open the door to new treatments for demyelinating diseases.


Asunto(s)
Enfermedades Desmielinizantes , Células Precursoras de Oligodendrocitos , Humanos , Enfermedades Desmielinizantes/patología , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Vaina de Mielina/metabolismo , Sistema Nervioso Central/metabolismo , Diferenciación Celular/fisiología , Antioxidantes , Receptores Acoplados a Proteínas G/metabolismo
3.
J Cell Biochem ; 124(4): 533-544, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36791278

RESUMEN

The human G protein coupled membrane receptor (GPR17), the sensor of brain damage, is identified as a biomarker for many neurological diseases. In human brain tissue, GPR17 exist in two isoforms, long and short. While cryo-electron microscopy technology has provided the structure of the long isoform of GPR17 with Gi complex, the structure of the short isoform and its activation mechanism remains unclear. Recently, we theoretically modeled the structure of the short isoform of GPR17 with Gi signaling protein and identified novel ligands. In the present work, we demonstrated the presence of two distinct ligand binding sites in the short isoform of GPR17. The molecular docking of GPR17 with endogenous (UDP) and synthetic ligands (T0510.3657, MDL29950) found the presence of two distinct binding pockets. Our observations revealed that endogenous ligand UDP can bind stronger in two different binding pockets as evidenced by glide and autodock vina scores, whereas the other two ligand's binding with GPR17 has less docking score. The analysis of receptor-UDP interactions shows complexes' stability in the lipid environment by 100 ns atomic molecular dynamics simulations. The amino acid residues VAL83, ARG87, and PHE111 constitute ligand binding site 1, whereas site 2 constitutes ASN67, ARG129, and LYS232. Root mean square fluctuation analysis showed the residues 83, 87, and 232 with higher fluctuations during molecular dynamics simulation in both binding pockets. Our findings imply that the residues of GPR17's two binding sites are crucial, and their interaction with UDP reveals the protein's hidden signaling and communication properties. Furthermore, this finding may assist in the development of targeted therapies for the treatment of neurological diseases.


Asunto(s)
Receptores Acoplados a Proteínas G , Uridina Difosfato , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Microscopía por Crioelectrón , Receptores Acoplados a Proteínas G/metabolismo , Sitios de Unión , Isoformas de Proteínas/metabolismo
4.
Neurobiol Dis ; 187: 106315, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37783234

RESUMEN

G protein-coupled receptor 17 (GPR17) and the WNT pathway are critical players of oligodendrocyte (OL) differentiation acting as essential timers in developing brain to achieve fully-myelinating cells. However, whether and how these two systems are related to each other is still unknown. Of interest, both factors are dysregulated in developing and adult brain diseases, including white matter injury and cancer, making the understanding of their reciprocal interactions of potential importance for identifying new targets and strategies for myelin repair. Here, by a combined pharmacological and biotechnological approach, we examined regulatory mechanisms linking WNT signaling to GPR17 expression in OLs. We first analyzed the relative expression of mRNAs encoding for GPR17 and the T cell factor/Lymphoid enhancer-binding factor-1 (TCF/LEF) transcription factors of the canonical WNT/ß-CATENIN pathway, in PDGFRα+ and O4+ OLs during mouse post-natal development. In O4+ cells, Gpr17 mRNA level peaked at post-natal day 14 and then decreased concomitantly to the physiological uprise of WNT tone, as shown by increased Lef1 mRNA level. The link between WNT signaling and GPR17 expression was further reinforced in vitro in primary PDGFRα+ cells and in Oli-neu cells. High WNT tone impaired OL differentiation and drastically reduced GPR17 mRNA and protein levels. In Oli-neu cells, WNT/ß-CATENIN activation repressed Gpr17 promoter activity through both putative WNT response elements (WRE) and upregulation of the inhibitor of DNA-binding protein 2 (Id2). We conclude that the WNT pathway influences OL maturation by repressing GPR17, which could have implications in pathologies characterized by dysregulations of the OL lineage including multiple sclerosis and oligodendroglioma.


Asunto(s)
Células Precursoras de Oligodendrocitos , Vía de Señalización Wnt , Ratones , Animales , beta Catenina/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Diferenciación Celular/fisiología , Oligodendroglía/metabolismo , ARN Mensajero/metabolismo
5.
J Neuroinflammation ; 20(1): 271, 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37990234

RESUMEN

BACKGROUND: Previously we reported that inhibition of GPR17 prevents amyloid ß 1-42 (Aß1-42)-induced cognitive impairment in mice. However, the role of GPR17 on cognition is still largely unknown. METHODS: Herein, we used a mouse model of cognitive impairment induced by lipopolysaccharide (LPS) to further investigate the role of GPR17 in cognition and its potential mechanism. The mice were pretreated with GPR17 shRNA lentivirus and cangrelor by microinjection into the dentate gyrus (DG) region of the hippocampus. After 21 days, LPS (0.25 mg/kg, i.p.) was administered for 7 days. Animal behavioral tests as well as pathological and biochemical assays were performed to evaluate the cognitive function in mice. RESULTS: LPS exposure resulted in a significant increase in GPR17 expression at both protein and mRNA levels in the hippocampus. Gene reduction and pharmacological blockade of GPR17 improved cognitive impairment in both the Morris water maze and novel object recognition tests. Knockdown and inhibition of GPR17 inhibited Aß production, decreased the expression of NF-κB p65, increased CREB phosphorylation and elevated BDNF expression, suppressed the accumulation of pro-inflammatory cytokines, inhibited Glial cells (microglia and astrocytes) activation, and increased Bcl-2, PSD-95, and SYN expression, reduced Bax expression as well as decreased caspase-3 activity and TUNEL-positive cells in the hippocampus of LPS-treated mice. Notably, knockdown and inhibition of GPR17 not only provided protective effects against cholinergic dysfunction but also facilitated the regulation of oxidative stress. In addition, cangrelor pretreatment can effectively inhibit the expression of inflammatory cytokines by suppressing NF-κB/CREB/BDNF signaling in BV-2 cells stimulated by LPS. However, activation of hippocampal GPR17 with MDL-29951 induced cognitive impairment in normal mice. CONCLUSIONS: These observations indicate that GPR17 may possess a neuroprotective effect against LPS-induced cognition deficits, and neuroinflammation by modulation of NF-κB/CREB/BDNF signaling in mice, indicating that GPR17 may be a promising new target for the prevention and treatment of AD.


Asunto(s)
Disfunción Cognitiva , Lipopolisacáridos , Ratones , Animales , Lipopolisacáridos/efectos adversos , FN-kappa B/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/prevención & control , Disfunción Cognitiva/metabolismo , Citocinas/metabolismo , Microglía/metabolismo , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
6.
Glia ; 70(6): 1052-1067, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35104015

RESUMEN

Physical activity (PA) promotes the proliferation of neural stem cells and enhances neurogenesis in the dentate gyrus resulting in hippocampal circuit remodeling and cognitive enhancement. Nonetheless, knowledge of other neural progenitors affected by PA and the mechanisms through which they could contribute to circuit plasticity and cognitive enhancement are still poorly understood. In this work we demonstrated that NG2-glia, also known as oligodendrocyte progenitor cells, show enhanced proliferation and differentiation in response to voluntary PA in a brain region-dependent manner in adult mice. Surprisingly, preventing NG2-glia differentiation during enhanced PA abolishes the exercise-associated cognitive improvement without affecting neurogenesis or baseline learning capacity. Thus, here we provided new evidence highlighting the requirement of oligodendrogenesis for exercise induced-cognition enhancement.


Asunto(s)
Células-Madre Neurales , Neurogénesis , Animales , Proliferación Celular/fisiología , Cognición/fisiología , Hipocampo , Ratones , Neurogénesis/fisiología
7.
Mol Ther ; 29(4): 1439-1458, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33309882

RESUMEN

Contrasting myelin damage through the generation of new myelinating oligodendrocytes represents a promising approach to promote functional recovery after stroke. Here, we asked whether activation of microglia and monocyte-derived macrophages affects the regenerative process sustained by G protein-coupled receptor 17 (GPR17)-expressing oligodendrocyte precursor cells (OPCs), a subpopulation of OPCs specifically reacting to ischemic injury. GPR17-iCreERT2:CAG-eGFP reporter mice were employed to trace the fate of GPR17-expressing OPCs, labeled by the green fluorescent protein (GFP), after permanent middle cerebral artery occlusion. By microglia/macrophages pharmacological depletion studies, we show that innate immune cells favor GFP+ OPC reaction and limit myelin damage early after injury, whereas they lose their pro-resolving capacity and acquire a dystrophic "senescent-like" phenotype at later stages. Intracerebral infusion of regenerative microglia-derived extracellular vesicles (EVs) restores protective microglia/macrophages functions, limiting their senescence during the post-stroke phase, and enhances the maturation of GFP+ OPCs at lesion borders, resulting in ameliorated neurological functionality. In vitro experiments show that EV-carried transmembrane tumor necrosis factor (tmTNF) mediates the pro-differentiating effects on OPCs, with future implications for regenerative therapies.


Asunto(s)
Senescencia Celular/genética , Vaina de Mielina/genética , Receptores Acoplados a Proteínas G/genética , Accidente Cerebrovascular/terapia , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Diferenciación Celular/genética , Línea Celular , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/terapia , Macrófagos/metabolismo , Macrófagos/trasplante , Masculino , Ratones , Microglía/metabolismo , Microglía/trasplante , Oligodendroglía/trasplante , Medicina Regenerativa/métodos , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Factor de Necrosis Tumoral alfa/genética
8.
Int J Mol Sci ; 24(1)2022 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-36613703

RESUMEN

GPR17, a G protein-coupled receptor, is a pivotal regulator of myelination. Its endogenous ligands trigger receptor desensitization and downregulation allowing oligodendrocyte terminal maturation. In addition to its endogenous agonists, GPR17 could be promiscuously activated by pro-inflammatory oxysterols and chemokines released at demyelinating lesions. Herein, the chemokine receptors CXCR2 and CXCR4 were selected to perform both in silico modelling and in vitro experiments to establish their structural and functional interactions with GPR17. The relative propensity of GPR17 and CXCR2 or CXCR4 to form homo- and hetero-dimers was assessed by homology modelling and molecular dynamics (MD) simulations, and co-immunoprecipitation and immunoenzymatic assay. The interaction between chemokine receptors and GPR17 was investigated by determining receptor-mediated modulation of intracellular cyclic adenosine monophosphate (cAMP). Our data show the GPR17 association with CXCR2 or CXCR4 and the negative regulation of these interactions by CXCR agonists or antagonists. Moreover, GPR17 and CXCR2 heterodimers can functionally influence each other. In contrast, CXCR4 can influence GPR17 functionality, but not vice versa. According to MD simulations, all the dimers reached conformational stability and negative formation energy, confirming the experimental observations. The cross-talk between these receptors could play a role in the development of the neuroinflammatory milieu associated with demyelinating events.


Asunto(s)
Receptores de Quimiocina , Receptores Acoplados a Proteínas G , Receptores Acoplados a Proteínas G/química , Transducción de Señal/fisiología , AMP Cíclico , Simulación de Dinámica Molecular
9.
Glia ; 69(7): 1709-1722, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33660902

RESUMEN

Oligodendroglial lineage cells go through a series of morphological changes before myelination. Prior to myelination, cell processes and membrane structures enlarge by approximately 7,000 times, which is required to support axonal wrapping and myelin segment formation. Failure of these processes leads to maldevelopment and impaired myelination. Quetiapine, an atypical antipsychotic drug, was proved to promote oligodendroglial differentiation and (re)myelination, pending detailed effects and regulatory mechanism. In this study, we showed that quetiapine promotes morphological maturation of oligodendroglial lineage cells and myelin segment formation, and a short-term quetiapine treatment is sufficient to induce these changes. To uncover the underlying mechanism, we examined the effect of quetiapine on the Oligodendrocyte transcription factor 1 (Olig1). We found that quetiapine upregulates Olig1 expression level and promotes nuclear Olig1 translocation to the cytosol, where it functions not as a transcription modulator, but in a way that highly correlates with oligodendrocyte morphological transformation. In addition, quetiapine treatment reverses the negative regulatory effect of the Olig1-regulated G protein-coupled receptor 17 (GPR17) on oligodendroglial morphological maturation. Our results demonstrate that quetiapine enhances oligodendroglial differentiation and myelination by promoting cell morphological transformation. This would shed light on the orchestration of oligodendroglia developmental mechanisms, and provides new targets for further therapeutic research.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Oligodendroglía , Axones/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Fumarato de Quetiapina/metabolismo , Fumarato de Quetiapina/farmacología
10.
Pflugers Arch ; 473(5): 775-783, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33712969

RESUMEN

White matter (WM) is a highly prominent feature in the human cerebrum and is comprised of bundles of myelinated axons that form the connectome of the brain. Myelin is formed by oligodendrocytes and is essential for rapid neuronal electrical communication that underlies the massive computing power of the human brain. Oligodendrocytes are generated throughout life by oligodendrocyte precursor cells (OPCs), which are identified by expression of the chondroitin sulphate proteoglycan NG2 (Cspg4), and are often termed NG2-glia. Adult NG2+ OPCs are slowly proliferating cells that have the stem cell-like property of self-renewal and differentiation into a pool of 'late OPCs' or 'differentiation committed' OPCs(COPs) identified by specific expression of the G-protein-coupled receptor GPR17, which are capable of differentiation into myelinating oligodendrocytes. In the adult brain, these reservoirs of OPCs and COPs ensure rapid myelination of new neuronal connections formed in response to neuronal signalling, which underpins learning and cognitive function. However, there is an age-related decline in myelination that is associated with a loss of neuronal function and cognitive decline. The underlying causes of myelin loss in ageing are manifold, but a key factor is the decay in OPC 'stemness' and a decline in their replenishment of COPs, which results in the ultimate failure of myelin regeneration. These changes in ageing OPCs are underpinned by dysregulation of neuronal signalling and OPC metabolic function. Here, we highlight the role of purine signalling in regulating OPC self-renewal and the potential importance of GPR17 and the P2X7 receptor subtype in age-related changes in OPC metabolism. Moreover, age is the main factor in the failure of myelination in chronic multiple sclerosis and myelin loss in Alzheimer's disease, hence understanding the importance of purine signalling in OPC regeneration and myelination is critical for developing new strategies for promoting repair in age-dependent neuropathology.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Oligodendroglía/metabolismo , Purinas/metabolismo , Animales , Axones/metabolismo , Axones/fisiología , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Humanos , Oligodendroglía/fisiología , Transducción de Señal
11.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34769111

RESUMEN

Characterization of new pharmacological targets is a promising approach in research of neurorepair mechanisms. The G protein-coupled receptor 17 (GPR17) has recently been proposed as an interesting pharmacological target, e.g., in neuroregenerative processes. Using the well-established ex vivo model of organotypic slice co-cultures of the mesocortical dopaminergic system (prefrontal cortex (PFC) and substantia nigra/ventral tegmental area (SN/VTA) complex), the influence of GPR17 ligands on neurite outgrowth from SN/VTA to the PFC was investigated. The growth-promoting effects of Montelukast (MTK; GPR17- and cysteinyl-leukotriene receptor antagonist), the glial cell line-derived neurotrophic factor (GDNF) and of two potent, selective GPR17 agonists (PSB-16484 and PSB-16282) were characterized. Treatment with MTK resulted in a significant increase in mean neurite density, comparable with the effects of GDNF. The combination of MTK and GPR17 agonist PSB-16484 significantly inhibited neuronal growth. qPCR studies revealed an MTK-induced elevated mRNA-expression of genes relevant for neuronal growth. Immunofluorescence labelling showed a marked expression of GPR17 on NG2-positive glia. Western blot and RT-qPCR analysis of untreated cultures suggest a time-dependent, injury-induced stimulation of GPR17. In conclusion, MTK was identified as a stimulator of neurite fibre outgrowth, mediating its effects through GPR17, highlighting GPR17 as an interesting therapeutic target in neuronal regeneration.


Asunto(s)
Acetatos/farmacología , Ciclopropanos/farmacología , Antagonistas de Leucotrieno/farmacología , Proyección Neuronal/efectos de los fármacos , Quinolinas/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Sulfuros/farmacología , Animales , Animales Recién Nacidos , Técnicas de Cocultivo , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Regeneración Nerviosa/efectos de los fármacos , Proyección Neuronal/genética , Ratas
12.
Glia ; 68(10): 1957-1967, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32086854

RESUMEN

Remyelination, namely, the formation of new myelin sheaths around denuded axons, counteracts axonal degeneration and restores neuronal function. Considerable advances have been made in understanding this regenerative process that often fails in diseases like multiple sclerosis, leaving axons demyelinated and vulnerable to damage, thus contributing to disease progression. The identification of the membrane receptor GPR17 on a subset of oligodendrocyte precursor cells (OPCs), which mediate remyelination in the adult central nervous system (CNS), has led to a huge amount of evidence that validated this receptor as a new attractive target for remyelinating therapies. Here, we summarize the role of GPR17 in OPC function, myelination and remyelination, describing its atypical pharmacology, its downstream signaling, and the genetic and epigenetic factors modulating its activity. We also highlight crucial insights into GPR17 pathophysiology coming from the demonstration that oligodendrocyte injury, associated with inflammation in chronic neurodegenerative conditions, is invariably characterized by abnormal and persistent GPR17 upregulation, which, in turn, is accompanied by a block of OPCs at immature premyelinating stages. Finally, we discuss the current literature in light of the potential exploitment of GPR17 as a therapeutic target to promote remyelination.


Asunto(s)
Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Remielinización/fisiología , Transducción de Señal/fisiología , Animales , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Epigénesis Genética/fisiología , Humanos , Vaina de Mielina/genética , Receptores Acoplados a Proteínas G/genética
13.
Int J Mol Sci ; 21(5)2020 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-32182666

RESUMEN

One of the most important goals in the treatment of demyelinating diseases such as multiple sclerosis (MS) is, in addition to immunomodulation, reconstruction of the lost myelin sheath. The modulator of the central nervous system myelination is the metabotropic receptor coupled to the G-protein: GPR17. GPR17 receptors are considered to be sensors of local damage to the myelin sheath, and play a role in the reconstruction and repair of demyelinating plaques caused by ongoing inflammatory processes. GPR17 receptors are present on nerve cells and precursor oligodendrocyte cells. Under physiological conditions, they are responsible for the differentiation and subsequent maturation of oligodendrocytes, while under pathological conditions (during damage to nerve cells), their expression increases to become mediators in the demyelinating processes. Moreover, they are essential not only in both the processes of inducing damage and the death of neurons, but also in the local repair of the damaged myelin sheath. Therefore, GPR17 receptors may be recognized as the potential goal in creating innovative therapies for the treatment of the neurodegenerative process in MS, based on the acceleration of the remyelination processes. This review examines the role of GRP17 in pathomechanisms of MS development.


Asunto(s)
Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Biomarcadores/metabolismo , Humanos , Esclerosis Múltiple/genética , Enfermedades Neurodegenerativas/genética , Receptores Acoplados a Proteínas G/genética , Remielinización/genética , Remielinización/fisiología
14.
Int J Mol Sci ; 21(7)2020 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-32244295

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive loss of motor neurons (MN). Importantly, MN degeneration is intimately linked to oligodendrocyte dysfunction and impaired capacity of oligodendrocyte precursor cells (OPCs) to regenerate the myelin sheath enwrapping and protecting neuronal axons. Thus, improving OPC reparative abilities represents an innovative approach to counteract MN loss. A pivotal regulator of OPC maturation is the P2Y-like G protein-coupled receptor 17 (GPR17), whose role in ALS has never been investigated. In other models of neurodegeneration, an abnormal increase of GPR17 has been invariably associated to myelin defects and its pharmacological manipulation succeeded in restoring endogenous remyelination. Here, we analyzed GPR17 alterations in the SOD1G93A ALS mouse model and assessed in vitro whether this receptor could be targeted to correct oligodendrocyte alterations. Western-blot and immunohistochemical analyses showed that GPR17 protein levels are significantly increased in spinal cord of ALS mice at pre-symptomatic stage; this alteration is exacerbated at late symptomatic phases. Concomitantly, mature oligodendrocytes degenerate and are not successfully replaced. Moreover, OPCs isolated from spinal cord of SOD1G93A mice display defective differentiation compared to control cells, which is rescued by treatment with the GPR17 antagonist montelukast. These data open novel therapeutic perspectives for ALS management.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Superóxido Dismutasa-1/metabolismo , Superóxido Dismutasa/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Vaina de Mielina/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Médula Espinal/metabolismo , Regulación hacia Arriba
15.
Proc Natl Acad Sci U S A ; 112(6): 1845-9, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25624481

RESUMEN

G protein-coupled receptor 17 (GPR17) was recently reported to be a Foxo1 target in agouti-related peptide (AGRP) neurons. Intracerebroventricular injection of GPR17 agonists induced food intake, whereas administration of an antagonist to the receptor reduced feeding. These data lead to the conclusion that pharmacological modulation of GPR17 has therapeutic potential to treat obesity. Here we report that mice deficient in Gpr17 (Gpr17(-/-)) have similar food intake and body weight compared with their wild-type littermates. Gpr17(-/-) mice have normal hypothalamic Agrp mRNA expression, AGRP plasma levels, and metabolic rate. GPR17 deficiency in mice did not affect glucose homeostasis or prevent fat-induced insulin resistance. These data do not support a role for GPR17 in the control of food intake, body weight, or glycemic control.


Asunto(s)
Ingestión de Alimentos/genética , Glucosa/metabolismo , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G/genética , Proteína Relacionada con Agouti/metabolismo , Análisis de Varianza , Animales , Secuencia de Bases , Composición Corporal/efectos de los fármacos , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neuronas/metabolismo , Análisis de Secuencia de ARN , Factores de Tiempo , Microtomografía por Rayos X
16.
J Neurosci ; 36(41): 10560-10573, 2016 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-27733608

RESUMEN

Demyelinating diseases, such as multiple sclerosis, are known to result from acute or chronic injury to the myelin sheath and inadequate remyelination; however, the underlying molecular mechanisms remain unclear. Here, we performed genome occupancy analysis by chromatin immunoprecipitation sequencing in oligodendrocytes in response to lysolecithin-induced injury and found that Olig2 and its downstream target Gpr17 are critical factors in regulating oligodendrocyte survival. After injury to oligodendrocytes, Olig2 was significantly upregulated and transcriptionally targeted the Gpr17 locus. Gpr17 activation inhibited oligodendrocyte survival by reducing the intracellular cAMP level and inducing expression of the pro-apoptotic gene Xaf1 The protein kinase A signaling pathway and the transcription factor c-Fos mediated the regulatory effects of Gpr17 in oligodendrocytes. We showed that Gpr17 inhibition elevated Epac1 expression and promoted oligodendrocyte differentiation. The loss of Gpr17, either globally or specifically in oligodendrocytes, led to an earlier onset of remyelination after myelin injury in mice. Similarly, pharmacological inhibition of Gpr17 with pranlukast promoted remyelination. Our findings indicate that Gpr17, an Olig2 transcriptional target, is activated after injury to oligodendrocytes and that targeted inhibition of Gpr17 promotes oligodendrocyte remyelination. SIGNIFICANCE STATEMENT: Genome occupancy analysis of oligodendrocytes in response to lysolecithin-mediated demyelination injury revealed that Olig2 and its downstream target Gpr17 are part of regulatory circuitry critical for oligodendrocyte survival. Gpr17 inhibits oligodendrocyte survival through activation of Xaf1 and cell differentiation by reducing Epac1 expression. The loss of Gpr17 in mice led to precocious myelination and an earlier onset of remyelination after demyelination. Pharmacological inhibition of Gpr17 promoted remyelination, highlighting the potential for Gpr17-targeted therapeutic approaches in demyelination diseases.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Supervivencia Celular/efectos de los fármacos , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Lisofosfatidilcolinas/toxicidad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Reguladoras de la Apoptosis , Diferenciación Celular/efectos de los fármacos , Cromonas/farmacología , Mapeo Cromosómico , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/efectos de los fármacos , Proteínas F-Box/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Factores de Intercambio de Guanina Nucleótido/genética , Antagonistas de Leucotrieno/farmacología , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción 2 de los Oligodendrocitos , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos
17.
J Biol Chem ; 291(2): 705-18, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26620557

RESUMEN

Recent studies have recognized G protein-coupled receptors as important regulators of oligodendrocyte development. GPR17, in particular, is an orphan G protein-coupled receptor that has been identified as oligodendroglial maturation inhibitor because its stimulation arrests primary mouse oligodendrocytes at a less differentiated stage. However, the intracellular signaling effectors transducing its activation remain poorly understood. Here, we use Oli-neu cells, an immortalized cell line derived from primary murine oligodendrocytes, and primary rat oligodendrocyte cultures as model systems to identify molecular targets that link cell surface GPR17 to oligodendrocyte maturation blockade. We demonstrate that stimulation of GPR17 by the small molecule agonist MDL29,951 (2-carboxy-4,6-dichloro-1H-indole-3-propionic acid) decreases myelin basic protein expression levels mainly by triggering the Gαi/o signaling pathway, which in turn leads to reduced activity of the downstream cascade adenylyl cyclase-cAMP-PKA-cAMP response element-binding protein (CREB). In addition, we show that GPR17 activation also diminishes myelin basic protein abundance by lessening stimulation of the exchange protein directly activated by cAMP (EPAC), thus uncovering a previously unrecognized role for EPAC to regulate oligodendrocyte differentiation. Together, our data establish PKA and EPAC as key downstream effectors of GPR17 that inhibit oligodendrocyte maturation. We envisage that treatments augmenting PKA and/or EPAC activity represent a beneficial approach for therapeutic enhancement of remyelination in those demyelinating diseases where GPR17 is highly expressed, such as multiple sclerosis.


Asunto(s)
Diferenciación Celular , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/citología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Factores de Intercambio de Guanina Nucleótido/metabolismo , Indoles/farmacología , Ratones , Modelos Biológicos , Proteína Básica de Mielina/metabolismo , Proteínas del Tejido Nervioso/agonistas , Fosforilación/efectos de los fármacos , Propionatos/farmacología , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/agonistas , Transducción de Señal , Tionucleótidos/farmacología
18.
Purinergic Signal ; 12(4): 661-672, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27544384

RESUMEN

Oligodendrocyte precursor cells (OPCs, also called NG2 cells) are scattered throughout brain parenchyma, where they function as a reservoir to replace lost or damaged oligodendrocytes, the myelin-forming cells. The hypothesis that, under some circumstances, OPCs can actually behave as multipotent cells, thus generating astrocytes and neurons as well, has arisen from some in vitro and in vivo evidence, but the molecular pathways controlling this alternative fate of OPCs are not fully understood. Their identification would open new opportunities for neuronal replace strategies, by fostering the intrinsic ability of the brain to regenerate. Here, we show that the anti-epileptic epigenetic modulator valproic acid (VPA) can promote the generation of new neurons from NG2+ OPCs under neurogenic protocols in vitro, through their initial de-differentiation to a stem cell-like phenotype that then evolves to "hybrid" cell population, showing OPC morphology but expressing the neuronal marker ßIII-tubulin and the GPR17 receptor, a key determinant in driving OPC transition towards myelinating oligodendrocytes. Under these conditions, the pharmacological blockade of the P2Y-like receptor GPR17 by cangrelor, a drug recently approved for human use, partially mimics the effects mediated by VPA thus accelerating cells' neurogenic conversion. These data show a co-localization between neuronal markers and GPR17 in vitro, and suggest that, besides its involvement in oligodendrogenesis, GPR17 can drive the fate of neural precursor cells by instructing precursors towards the neuronal lineage. Being a membrane receptor, GPR17 represents an ideal "druggable" target to be exploited for innovative regenerative approaches to acute and chronic brain diseases.


Asunto(s)
Células Madre Multipotentes/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Linaje de la Célula/fisiología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Células Madre Multipotentes/citología , Células-Madre Neurales/citología , Neuronas/citología , Oligodendroglía/citología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética
19.
Glia ; 63(12): 2327-39, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26228571

RESUMEN

During oligodendrocyte precursor cell (OPC) differentiation, defective control of the membrane receptor GPR17 has been suggested to block cell maturation and impair remyelination under demyelinating conditions. After the immature oligodendrocyte stage, to enable cells to complete maturation, GPR17 is physiologically down-regulated via phosphorylation/desensitization by G protein-coupled receptor kinases (GRKs); conversely, GRKs are regulated by the "mammalian target of rapamycin" mTOR. However, how GRKs and mTOR are connected to each other in modulating GPR17 function and oligodendrogenesis has remained elusive. Here we show, for the first time, a role for Murine double minute 2 (Mdm2), a ligase previously involved in ubiquitination/degradation of the onco-suppressor p53 protein. In maturing OPCs, both rapamycin and Nutlin-3, a small molecule inhibitor of Mdm2-p53 interactions, increased GRK2 sequestration by Mdm2, leading to impaired GPR17 down-regulation and OPC maturation block. Thus, Mdm2 intertwines mTOR with GRK2 in regulating GPR17 and oligodendrogenesis and represents a novel actor in myelination.


Asunto(s)
Oligodendroglía/fisiología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Animales , Células Cultivadas , Fármacos del Sistema Nervioso Central/farmacología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiología , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Imidazoles/farmacología , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/efectos de los fármacos , Piperazinas/farmacología , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
20.
J Cell Mol Med ; 18(9): 1785-96, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24909956

RESUMEN

GPR17 is a G(i) -coupled dual receptor activated by uracil-nucleotides and cysteinyl-leukotrienes. These mediators are massively released into hypoxic tissues. In the normal heart, GPR17 expression has been reported. By contrast, its role in myocardial ischaemia has not yet been assessed. In the present report, the expression of GPR17 was investigated in mice before and at early stages after myocardial infarction by using immunofluorescence, flow cytometry and RT-PCR. Before induction of ischaemia, results indicated the presence of the receptor in a population of stromal cells expressing the stem-cell antigen-1 (Sca-1). At early stages after ligation of the coronary artery, the receptor was expressed in Sca-1(+) cells, and cells stained with Isolectin-B4 and anti-CD45 antibody. GPR17(+) cells also expressed mesenchymal marker CD44. GPR17 function was investigated in vitro in a Sca-1(+)/CD31(-) cell line derived from normal hearts. These experiments showed a migratory function of the receptor by treatment with UDP-glucose and leukotriene LTD4, two GPR17 pharmacological agonists. The GPR17 function was finally assessed in vivo by treating infarcted mice with Cangrelor, a pharmacological receptor antagonist, which, at least in part, inhibited early recruitment of GPR17(+) and CD45(+) cells. These findings suggest a regulation of heart-resident mesenchymal cells and blood-borne cellular species recruitment following myocardial infarction, orchestrated by GPR17.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Infarto del Miocardio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Animales , Antígenos Ly/metabolismo , Movimiento Celular , Receptores de Hialuranos , Antígenos Comunes de Leucocito/metabolismo , Leucotrieno D4/farmacología , Leucotrieno D4/fisiología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Proteínas del Tejido Nervioso/agonistas , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Acoplados a Proteínas G/agonistas , Uridina Difosfato Glucosa/farmacología , Uridina Difosfato Glucosa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA