Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Intervalo de año de publicación
1.
Neuropharmacology ; 247: 109850, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38295947

RESUMEN

Adolescence is a critical period for brain maturation in which this organ undergoes critical plasticity mechanisms that increase its vulnerability to the effects of alcohol. Significantly, ethanol-induced disruption of hippocampal neurogenesis has been related to cognitive decline in adulthood. During adolescence, the maturation of perineuronal nets (PNNs), extracellular matrix structures highly affected by ethanol consumption, plays a fundamental role in neurogenesis and plasticity in the hippocampus. Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ is a critical anchor point for PNNs on the cell surface. Using the adolescent intermittent access to ethanol (IAE) model, we previously showed that MY10, a small-molecule inhibitor of RPTPß/ζ, reduces chronic ethanol consumption in adolescent male mice but not in females and prevents IAE-induced neurogenic loss in the male hippocampus. We have now tested if these effects of MY10 are related to sex-dependent modulatory actions on ethanol-induced effects in PNNs. Our findings suggest a complex interplay between alcohol exposure, neural structures, and sex-related differences in the modulation of PNNs and parvalbumin (PV)-positive cells in the hippocampus. In general, IAE increased the number of PV + cells in the female hippocampus and reduced PNNs intensity in different hippocampal regions, particularly in male mice. Notably, we found that pharmacological inhibition of RPTPß/ζ with MY10 regulates ethanol-induced alterations of PNNs intensity, which correlates with the protection of hippocampal neurogenesis from ethanol neurotoxic effects and may be related to the capacity of MY10 to increase the gene expression of key components of PNNs.


Asunto(s)
Etanol , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores , Ratones , Masculino , Animales , Femenino , Etanol/farmacología , Etanol/metabolismo , Matriz Extracelular/metabolismo , Hipocampo/metabolismo , Consumo de Bebidas Alcohólicas
2.
Int J Cancer ; 153(5): 1051-1066, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37260355

RESUMEN

Protein tyrosine phosphatase receptor zeta 1 (PTPRZ1) is a transmembrane tyrosine phosphatase (TP) expressed in endothelial cells and required for stimulation of cell migration by vascular endothelial growth factor A165 (VEGFA165 ) and pleiotrophin (PTN). It is also over or under-expressed in various tumor types. In this study, we used genetically engineered Ptprz1-/- and Ptprz1+/+ mice to study mechanistic aspects of PTPRZ1 involvement in angiogenesis and investigate its role in lung adenocarcinoma (LUAD) growth. Ptprz1-/- lung microvascular endothelial cells (LMVEC) have increased angiogenic features compared with Ptprz1+/+ LMVEC, in line with the increased lung angiogenesis and the enhanced chemically induced LUAD growth in Ptprz1-/- compared with Ptprz1+/+ mice. In LUAD cells isolated from the lungs of urethane-treated mice, PTPRZ1 TP inhibition also enhanced proliferation and migration. Expression of beta 3 (ß3 ) integrin is decreased in Ptprz1-/- LMVEC, linked to enhanced VEGF receptor 2 (VEGFR2), c-Met tyrosine kinase (TK) and Akt kinase activities. However, only c-Met and Akt seem responsible for the enhanced endothelial cell activation in vitro and LUAD growth and angiogenesis in vivo in Ptprz1-/- mice. A selective PTPRZ1 TP inhibitor, VEGFA165 and PTN also activate c-Met and Akt in a PTPRZ1-dependent manner in endothelial cells, and their stimulatory effects are abolished by the c-Met TK inhibitor (TKI) crizotinib. Altogether, our data suggest that low PTPRZ1 expression is linked to worse LUAD prognosis and response to c-Met TKIs and uncover for the first time the role of PTPRZ1 in mediating c-Met activation by VEGFA and PTN.


Asunto(s)
Adenocarcinoma del Pulmón , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores , Animales , Ratones , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Células Endoteliales/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tirosina/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo
3.
Biomedicines ; 11(5)2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37238989

RESUMEN

Binge drinking during adolescence increases the risk of alcohol use disorder, possibly by involving alterations of neuroimmune responses. Pleiotrophin (PTN) is a cytokine that inhibits Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ. PTN and MY10, an RPTPß/ζ pharmacological inhibitor, modulate ethanol behavioral and microglial responses in adult mice. Now, to study the contribution of endogenous PTN and the implication of its receptor RPTPß/ζ in the neuroinflammatory response in the prefrontal cortex (PFC) after acute ethanol exposure in adolescence, we used MY10 (60 mg/kg) treatment and mice with transgenic PTN overexpression in the brain. Cytokine levels by X-MAP technology and gene expression of neuroinflammatory markers were determined 18 h after ethanol administration (6 g/kg) and compared with determinations performed 18 h after LPS administration (5 g/kg). Our data indicate that Ccl2, Il6, and Tnfa play important roles as mediators of PTN modulatory actions on the effects of ethanol in the adolescent PFC. The data suggest PTN and RPTPß/ζ as targets to differentially modulate neuroinflammation in different contexts. In this regard, we identified for the first time important sex differences that affect the ability of the PTN/RPTPß/ζ signaling pathway to modulate ethanol and LPS actions in the adolescent mouse brain.

4.
Neuropharmacology ; 227: 109438, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36706907

RESUMEN

Pleiotrophin (PTN) is a cytokine that modulates ethanol drinking and reward and regulates glial responses in different contexts. PTN is an inhibitor of Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ. Inhibition of RPTPß/ζ reduces binge-like drinking in adult male mice. Whether inhibition of RPTPß/ζ is effective in reducing ethanol consumption during adolescence and in both sexes remained to be studied. In this work, male and female adolescent mice underwent an intermittent access to ethanol (IAE) 2-bottle choice protocol. Treatment with MY10 (60 mg/kg, i.g.), a small-molecule RPTPß/ζ inhibitor, reduced chronic 3-week ethanol consumption only in male mice. We detected an ethanol-induced overall decrease in hippocampal GFAPir and Iba1ir, independently of the treatment received, suggesting that RPTPß/ζ is not key in the regulation of IAE-induced glial responses. However, we found a significant negative correlation between the size of microglial cells and the number of hippocampal neuronal progenitors only in male mice after IAE. This correlation was disrupted by treatment with MY10 before each drinking session, which may be related to the ability of MY10 to regulate the intensity of the perineuronal nets (PNNs) in the hippocampus in a sex-dependent manner. The data show for the first time that inhibition of RPTPß/ζ reduces chronic voluntary ethanol consumption in adolescent mice in a sex-dependent manner. In addition, we show evidence for sex-specific differences in the effects of IAE on glial responses and hippocampal neurogenesis, which may be related to different actions of the RPTPß/ζ signalling pathway in the brains of male and female mice.


Asunto(s)
Etanol , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores , Femenino , Ratones , Masculino , Animales , Etanol/farmacología , Transducción de Señal , Neuroglía/metabolismo , Citocinas/metabolismo , Neurogénesis
5.
Neurotoxicology ; 94: 98-107, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36402194

RESUMEN

Adolescence is a critical period for brain maturation in which this organ is more vulnerable to the damaging effects of ethanol. Administration of ethanol in mice induces a rapid cerebral upregulation of pleiotrophin (PTN), a cytokine that regulates the neuroinflammatory processes induced by different insults and the behavioral effects of ethanol. PTN binds Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ and inhibits its phosphatase activity, suggesting that RPTPß/ζ may be involved in the regulation of ethanol effects. To test this hypothesis, we have treated adolescent mice with the RPTPß/ζ inhibitor MY10 (60 mg/kg) before an acute ethanol (6 g/kg) administration. Treatment with MY10 completely prevented the ethanol-induced neurogenic loss in the hippocampus of both male and female mice. In flow cytometry studies, ethanol tended to increase the number of NeuN+/activated Caspase-3+ cells particularly in female mice, but no significant effects were found. Ethanol increased Iba1+ cell area and the total marked area in the hippocampus of female mice, suggesting sex differences in ethanol-induced microgliosis. In addition, ethanol reduced the circulating levels of IL-6 and IL-10 in both sexes, although this reduction was only found significant in males and not affected by MY10 treatment. Interestingly, MY10 alone increased the total marked area and the number of Iba1+ cells only in the female hippocampus, but tended to reduce the circulating levels of TNF-α only in male mice. In summary, the data identify a novel modulatory role of RPTPß/ζ on ethanol-induced loss of hippocampal neurogenesis, which seems unrelated to glial and inflammatory responses. The data also suggest sex differences in RPTPß/ζ function that may be relevant to immune responses and ethanol-induced microglial responses.


Asunto(s)
Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores , Transducción de Señal , Animales , Femenino , Masculino , Ratones , Citocinas/metabolismo , Etanol/toxicidad , Hipocampo/metabolismo , Neurogénesis , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo
6.
Sci Rep ; 12(1): 3577, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35246557

RESUMEN

Pleiotrophin (PTN) is a cytokine involved in nerve tissue repair processes, neuroinflammation and neuronal survival. PTN expression levels are upregulated in the nigrostriatal pathway of Parkinson's Disease (PD) patients. We aimed to characterize the dopaminergic injury and glial responses in the nigrostriatal pathway of mice with transgenic Ptn overexpression in the brain (Ptn-Tg) after intrastriatal injection of the catecholaminergic toxic 6-hydroxydopamine (6-OHDA) at a low dose (5 µg). Ten days after surgery, the injection of 6-OHDA induced a significant decrease of the number of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra and of the striatal TH contents in Wild type (Wt) mice. In contrast, these effects of 6-OHDA were absent in Ptn-Tg mice. When the striatal Iba1 and GFAP immunoreactivity was studied, no statistical differences were found between vehicle-injected Wt and Ptn-Tg mice. Furthermore, 6-OHDA did not cause robust glial responses neither on Wt or Ptn-Tg mice 10 days after injections. In metabolomics studies, we detected interesting metabolites that significantly discriminate the more injured 6-OHDA-injected Wt striatum and the more protected 6-OHDA-injected Ptn-Tg striatum. Particularly, we detected groups of metabolites, mostly corresponding to phospholipids, whose trends were opposite in both groups. In summary, the data confirm lower 6-OHDA-induced decreases of TH contents in the nigrostriatal pathway of Ptn-Tg mice, suggesting a neuroprotective effect of brain PTN overexpression in this mouse model of PD. New lipid-related PD drug candidates emerge from this study and the data presented here support the increasingly recognized "lipid cascade" in PD.


Asunto(s)
Enfermedad de Parkinson , Animales , Proteínas Portadoras , Cuerpo Estriado/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Lípidos/farmacología , Metabolómica , Ratones , Oxidopamina/farmacología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
7.
Neurosci Lett ; 762: 136156, 2021 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-34358624

RESUMEN

Pleiotrophin (PTN) and midkine (MK) are growth factors that modulate alcohol consumption and reward. Since both PTN and MK limit the rewarding effects of alcohol, pharmacological potentiation of the PTN and MK signaling pathways has been proposed for the treatment of alcohol use disorders (AUD). Although the use of this therapy in the prevention of alcohol relapse is important, the potential role of these cytokines in extinguishing alcohol-induced seeking behavior is a key question that remains unanswered. To fill this gap, we have now studied the extinction of the conditioned place preference (CPP) induced by different doses of alcohol in Ptn knockout (Ptn-/-) and Mk knockout (Mk-/-) mice. The data confirm a higher sensitivity of Ptn-/- mice to the conditioning effects of a low dose (1 g/kg) and a rewarding dose (2 g/kg) of alcohol, while Mk-/- mice are only more susceptible to the conditioning effects of the low dose of this drug. More importantly, the percentage of Mk-/- mice, not Ptn-/- mice, that efficiently extinguished alcohol-induced CPP was significantly higher than that of Wt mice. Taken together, the data presented here confirm that Ptn and Mk are genetic factors that determine the conditioning effects of alcohol in mice and that Mk is a novel factor that plays an important role in the extinction of alcohol-induced CPP.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Alcoholismo/genética , Condicionamiento Clásico/efectos de los fármacos , Extinción Psicológica/fisiología , Midkina/metabolismo , Animales , Proteínas Portadoras/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Citocinas/metabolismo , Comportamiento de Búsqueda de Drogas/fisiología , Etanol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
8.
Int J Mol Sci ; 22(13)2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206170

RESUMEN

Pleiotrophin (PTN) is a neurotrophic factor that regulates glial responses in animal models of different types of central nervous system (CNS) injuries. PTN is upregulated in the brain in different pathologies characterized by exacerbated neuroinflammation, including Parkinson's disease. PTN is an endogenous inhibitor of Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ, which is abundantly expressed in the CNS. Using a specific inhibitor of RPTPß/ζ (MY10), we aimed to assess whether the PTN/RPTPß/ζ axis is involved in neuronal and glial injury induced by the toxin MPP+. Treatment with the RPTPß/ζ inhibitor MY10 alone decreased the viability of both SH-SY5Y neuroblastoma cells and BV2 microglial cultures, suggesting that normal RPTPß/ζ function is involved in neuronal and microglial viability. We observed that PTN partially decreased the cytotoxicity induced by MPP+ in SH-SY5Y cells underpinning the neuroprotective function of PTN. However, MY10 did not seem to modulate the SH-SY5Y cell loss induced by MPP+. Interestingly, we observed that media from SH-SY5Y cells treated with MPP+ and MY10 decreases microglial viability but may elicit a neuroprotective response of microglia by upregulating Ptn expression. The data suggest a neurotrophic role of microglia in response to neuronal injury through upregulation of Ptn levels.


Asunto(s)
Proteínas Portadoras/metabolismo , Comunicación Celular , Citocinas/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Animales , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Ratones , Microglía/fisiología , Modelos Biológicos , Neuronas/fisiología , Enfermedad de Parkinson/fisiopatología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/fisiología , Transducción de Señal
9.
Sci Rep ; 10(1): 20259, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33219280

RESUMEN

Pleiotrophin (PTN) is a cytokine that is upregulated in different neuroinflammatory disorders. Using mice with transgenic PTN overexpression in the brain (Ptn-Tg), we have found a positive correlation between iNos and Tnfα mRNA and Ptn mRNA levels in the prefrontal cortex (PFC) of LPS-treated mice. PTN is an inhibitor of Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ, which is mainly expressed in the central nervous system. We aimed to test if RPTPß/ζ is involved in the modulation of neuroinflammatory responses using specific inhibitors of RPTPß/ζ (MY10 and MY33-3). Treatment with MY10 potentiated LPS-induced microglial responses in the mouse PFC. Surprisingly, MY10 caused a decrease in LPS-induced NF-κB p65 expression, suggesting that RPTPß/ζ may be involved in a novel mechanism of potentiation of microglial activation independent of the NF-κB p65 pathway. MY33-3 and MY10 limited LPS-induced nitrites production and iNos increases in BV2 microglial cells. SH-SY5Y neuronal cells were treated with the conditioned media from MY10/LPS-treated BV2 cells. Conditioned media from non-stimulated and from LPS-stimulated BV2 cells increased the viability of SH-SY5Y cultures. RPTPß/ζ inhibition in microglial cells disrupted this neurotrophic effect of microglia, suggesting that RPTPß/ζ plays a role in the neurotrophic phenotype of microglia and in microglia-neuron communication.


Asunto(s)
Comunicación Celular/fisiología , Microglía/citología , Neuronas/citología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/fisiología , Animales , Proteínas Portadoras/genética , Citocinas/genética , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
10.
Behav Brain Res ; 369: 111933, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31054277

RESUMEN

Pleiotrophin (PTN) and Midkine (MK) are neurotrophic factors that are upregulated in the prefrontal cortex after alcohol administration and have been shown to reduce ethanol drinking and reward. PTN and MK are endogenous inhibitors of Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ. Interestingly, pharmacological inhibition of RPTPß/ζ reduces ethanol consumption and blocks ethanol-induced conditioned place preference (CPP) in wild type mice. Since PTN-knockout (Ptn-/-) mice are more sensitive to the conditioning effects of alcohol, we aimed to test the effects of MY10, a small-molecule inhibitor of RPTPß/ζ, on ethanol-induced CPP in Ptn-/- mice. The data presented here demonstrate for the first time that a regular dose of MY10, known to block ethanol consumption and reward in wild type mice, also blocks the rewarding effects of ethanol in the more vulnerable individuals lacking PTN, the endogenous inhibitor of RPTPß/ζ. In addition, since MY10 readily penetrates the blood brain barrier (BBB), we tested its effects in a series of behavioural tests in Ptn+/+ and Ptn-/- mice. The data indicate that MY10 does not cause gross behavioural effects in wild type mice. However, MY10 tended to induce anxiolytic effects in Ptn-/- mice in the elevated plus maze paradigm. Overall, the data indicate that MY10 rescues Ptn-/- mice from their increased susceptibility to the conditioning effects of ethanol and may induce anxiolytic effects in individuals with reduced or absent PTN functions. Further studies are needed to confirm the potential of pharmacological inhibition of RPTPß/ζ as a new therapeutic strategy in the treatment of anxiety-related disorders.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Condicionamiento Clásico/efectos de los fármacos , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/antagonistas & inhibidores , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Citocinas/genética , Citocinas/metabolismo , Etanol/metabolismo , Etanol/farmacología , Inhibición Psicológica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Midkina/genética , Midkina/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Recompensa , Transducción de Señal/efectos de los fármacos
11.
Front Pharmacol ; 10: 377, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31031625

RESUMEN

Inflammation is a common factor of pathologies such as obesity, type 2 diabetes or neurodegenerative diseases. Chronic inflammation is considered part of the pathogenic mechanisms of different disorders associated with aging. Interestingly, peripheral inflammation and the associated metabolic alterations not only facilitate insulin resistance and diabetes but also neurodegenerative disorders. Therefore, the identification of novel pathways, common to the development of these diseases, which modulate the immune response and signaling is key. It will provide highly relevant information to advance our knowledge of the multifactorial process of aging, and to establish new biomarkers and/or therapeutic targets to counteract the underlying chronic inflammatory processes. One novel pathway that regulates peripheral and central immune responses is triggered by the cytokines pleiotrophin (PTN) and midkine (MK), which bind its receptor, Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ, and inactivate its phosphatase activity. In this review, we compile a growing body of knowledge suggesting that PTN and MK modulate the immune response and/or inflammation in different pathologies characterized by peripheral inflammation associated with insulin resistance, such as aging, and in central disorders characterized by overt neuroinflammation, such as neurodegenerative diseases and endotoxemia. Evidence strongly suggests that regulation of the PTN and MK signaling pathways may provide new therapeutic opportunities particularly in those neurological disorders characterized by increased PTN and/or MK cerebral levels and neuroinflammation. Importantly, we discuss existing therapeutics, and others being developed, that modulate these signaling pathways, and their potential use in pathologies characterized by overt neuroinflammation.

12.
Diabetologia ; 62(1): 123-135, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30327824

RESUMEN

AIMS/HYPOTHESIS: Pleiotrophin, a developmentally regulated and highly conserved cytokine, exerts different functions including regulation of cell growth and survival. Here, we hypothesise that this cytokine can play a regulatory role in glucose and lipid homeostasis. METHODS: To test this hypothesis, we performed a longitudinal study characterising the metabolic profile (circulating variables and tissue mRNA expression) of gene-targeted Ptn-deficient female mice and their corresponding wild-type counterparts at different ages from young adulthood (3 months) to older age (15 months). Metabolic cages were used to investigate the respiratory exchange ratio and energy expenditure, at both 24°C and 30°C. Undifferentiated immortalised mouse brown adipocytes (mBAs) were treated with 0.1 µg/ml pleiotrophin until day 6 of differentiation, and markers of mBA differentiation were analysed by quantitative real-time PCR (qPCR). RESULTS: Ptn deletion was associated with a reduction in total body fat (20.2% in Ptn+/+ vs 13.9% in Ptn-/- mice) and an enhanced lipolytic response to isoprenaline in isolated adipocytes from 15-month-old mice (189% in Ptn+/+ vs 273% in Ptn-/- mice). We found that Ptn-/- mice exhibited a significantly lower QUICKI value and an altered lipid profile; plasma triacylglycerols and NEFA did not increase with age, as happens in Ptn+/+ mice. Furthermore, the contribution of cold-induced thermogenesis to energy expenditure was greater in Ptn-/- than Ptn+/+ mice (42.6% and 33.6%, respectively). Body temperature and the activity and expression of deiodinase, T3 and mitochondrial uncoupling protein-1 in the brown adipose tissue of Ptn-/- mice were higher than in wild-type controls. Finally, supplementing brown pre-adipocytes with pleiotrophin decreased the expression of the brown adipocyte markers Cidea (20% reduction), Prdm16 (21% reduction), and Pgc1-α (also known as Ppargc1a, 11% reduction). CONCLUSIONS/INTERPRETATION: Our results reveal for the first time that pleiotrophin is a key player in preserving insulin sensitivity, driving the dynamics of adipose tissue lipid turnover and plasticity, and regulating energy metabolism and thermogenesis. These findings open therapeutic avenues for the treatment of metabolic disorders by targeting pleiotrophin in the crosstalk between white and brown adipose tissue.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Metabolismo Energético/fisiología , Termogénesis/fisiología , Animales , Proteínas Portadoras/genética , Citocinas/genética , Metabolismo Energético/genética , Femenino , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Estudios Longitudinales , Ratones , Ratones Noqueados , Termogénesis/genética
13.
Neuropharmacology ; 137: 86-95, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29753117

RESUMEN

Pleiotrophin (PTN) and Midkine (MK) are neurotrophic factors that are upregulated in the prefrontal cortex after alcohol administration and have been shown to reduce ethanol drinking and reward. PTN and MK are the endogenous inhibitors of Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ (a.k.a. PTPRZ1, RPTPß, PTPζ), suggesting a potential role for this phosphatase in the regulation of alcohol effects. To determine if RPTPß/ζ regulates ethanol consumption, we treated mice with recently developed small-molecule inhibitors of RPTPß/ζ (MY10, MY33-3) before testing them for binge-like drinking using the drinking in the dark protocol. Mice treated with RPTPß/ζ inhibitors, particularly with MY10, drank less ethanol than controls. MY10 treatment blocked ethanol conditioned place preference, showed limited effects on ethanol-induced ataxia, and potentiated the sedative effects of ethanol. We also tested whether RPTPß/ζ is involved in ethanol signaling pathways. We found that ethanol treatment of neuroblastoma cells increased phosphorylation of anaplastic lymphoma kinase (ALK) and TrkA, known substrates of RPTPß/ζ. Treatment of neuroblastoma cells with MY10 or MY33-3 also increased levels of phosphorylated ALK and TrkA. However, concomitant treatment of neuroblastoma cells with ethanol and MY10 or MY33-3 prevented the increase in pTrkA and pALK. These results demonstrate for the first time that ethanol engages TrkA signaling and that RPTPß/ζ modulates signaling pathways activated by alcohol and behavioral responses to this drug. The data support the hypothesis that RPTPß/ζ might be a novel target of pharmacotherapy for reducing excessive alcohol consumption.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/enzimología , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/antagonistas & inhibidores , Disuasivos de Alcohol/síntesis química , Disuasivos de Alcohol/química , Disuasivos de Alcohol/farmacología , Quinasa de Linfoma Anaplásico/metabolismo , Animales , Consumo Excesivo de Bebidas Alcohólicas/tratamiento farmacológico , Línea Celular Tumoral , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Ratones Endogámicos C57BL , Receptor trkA/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo
14.
Eur J Med Chem ; 144: 318-329, 2018 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-29275231

RESUMEN

A new series of blood-brain barrier permeable molecules designed to mimic the activity of Pleiotrophin in the CNS has been designed and synthesized. These compounds exert their action by interacting with the intracellular domain PD1 of the Protein Tyrosine-Phosphatase Receptor Z1 (PTPRZ1), and inhibiting its tyrosine phosphatase activity. The most potent compounds 10a and 12b (IC50 = 0,1 µM) significantly increase the phosphorylation of key tyrosine residues of PTPRZ1 substrates involved in neuronal survival and differentiation, and display protective effects against amphetamine-induced toxicity. Docking and molecular dynamics experiments have been used to analyze the binding mode and to explain the observed selectivity against PTP1B. An In vivo experiment has demonstrated that 10a can cross the BBB, thus promoting the possibility of moving forward these candidates for the development of drugs for the treatment of CNS disorders, such as drug addiction and neurodegenerative diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Proteínas Portadoras/farmacología , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Citocinas/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/antagonistas & inhibidores , Animales , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/química , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Proteínas Portadoras/síntesis química , Proteínas Portadoras/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Enfermedades del Sistema Nervioso Central/metabolismo , Citocinas/síntesis química , Citocinas/química , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Ratones , Modelos Moleculares , Estructura Molecular , Ratas , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Relación Estructura-Actividad
15.
Neurosci Lett ; 662: 213-218, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29061398

RESUMEN

Pleiotrophin (PTN) and Midkine (MK) are two growth factors that modulate neuroinflammation. PTN overexpression in the brain prevents LPS-induced astrocytosis in mice but potentiates microglial activation. The modest astrocytic response caused by a low dose of LPS (0.5mg/kg) is blocked in the striatum of MK-/- mice whereas microglial response is unaffected. We have now tested the effects of an intermediate dose of LPS (7.5mg/kg) in glial response in PTN-/- and MK-/- mice. We found that LPS-induced astrocytosis is prevented in prefrontal cortex and striatum of both PTN-/- and MK-/- mice. Some of the morphological changes of microglia induced by LPS tended to increase in both genotypes, particularly in PTN-/- mice. Since we previously showed that PTN potentiates LPS-induced activation of BV2 microglial cells, we tested the activation of FYN kinase, a substrate of the PTN receptor RPTPß/ζ, and the subsequent ERK1/2 phosphorylation on LPS and PTN-treated BV2 cells. LPS effects on BV2 cells were not affected by the addition of PTN, suggesting that PTN does not recruit the FYN-MAP kinase signaling pathway in order to modulate LPS effects on microglial cells. Taking together, evidences demonstrate that regulation of astroglial responses to LPS administration are highly dependent on the levels of expression of PTN and MK. Further studies are needed to clarify the possible roles of endogenous expression of PTN and MK in LPS-induced microglial responses.


Asunto(s)
Astrocitos/metabolismo , Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Encefalitis/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lipopolisacáridos/administración & dosificación , Microglía/metabolismo , Animales , Astrocitos/efectos de los fármacos , Línea Celular , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Encefalitis/inducido químicamente , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Midkina , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Transducción de Señal/efectos de los fármacos
16.
J Neuroinflammation ; 14(1): 46, 2017 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-28259175

RESUMEN

BACKGROUND: Pleiotrophin (PTN) is a cytokine found highly upregulated in the brain in different disorders characterized by overt neuroinflammation such as neurodegenerative diseases, drug addiction, traumatic injury, and ischemia. In the present work, we have explored whether PTN modulates neuroinflammation and if Toll-like receptor 4 (TLR4), crucial in the initiation of an immune response, is involved. METHODS: In immunohistochemistry assays, we studied lipopolysaccharide (LPS, 7.5 mg/kg i.p.)-induced changes in glial fibrillary acidic protein (GFAP, astrocyte marker) and ionized calcium-binding adaptor molecule 1 (Iba1, microglia marker) expression in the prefrontal cortex (PFC) and striatum of mice with transgenic PTN overexpression in the brain (PTN-Tg) and in wild-type (WT) mice. Cytokine protein levels were assessed in the PFC by X-MAP technology. The influence of TLR4 signaling in LPS effects in both genotypes was assessed by pretreatment with the TLR4 antagonist (TAK-242, 3.0 mg/kg i.p.). Murine BV2 microglial cells were treated with PTN (0.5 µg/ml) and LPS (1.0 µg/ml) and assessed for the release of nitric oxide (NO). RESULTS: We found that LPS-induced microglial activation is significantly increased in the PFC of PTN-Tg mice compared to that of WT mice. The levels of TNF-α, IL-6, and MCP-1 in response to LPS were significantly increased in the PFC of PTN-Tg mice compared to that of WT mice. Pretreatment with TAK-242 efficiently blocked increases in cytokine contents in a similar manner in both genotypes. Concomitant incubation of BV2 cells with LPS and PTN significantly potentiated the production of NO compared to cells only treated with LPS. CONCLUSIONS: Our findings identify for the first time that PTN is a novel and potent regulator of neuroinflammation. Pleiotrophin potentiates LPS-stimulated microglia activation. Our results suggest that regulation of the PTN signaling pathways may constitute new therapeutic opportunities particularly in those neurological disorders characterized by increased PTN cerebral levels and neuroinflammation.


Asunto(s)
Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Encefalitis/patología , Microglía/fisiología , Análisis de Varianza , Animales , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/genética , Línea Celular Transformada , Citocinas/genética , Relación Dosis-Respuesta a Droga , Encefalitis/inducido químicamente , Encefalitis/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Óxido Nítrico/metabolismo , Corteza Prefrontal/patología , Sulfonamidas/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/metabolismo
17.
Eur Neuropsychopharmacol ; 26(11): 1794-1805, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27642078

RESUMEN

It was previously shown that mice with genetic deletion of the neurotrophic factor pleiotrophin (PTN-/-) show enhanced amphetamine neurotoxicity and impair extinction of amphetamine conditioned place preference (CPP), suggesting a modulatory role of PTN in amphetamine neurotoxicity and reward. We have now studied the effects of amphetamine (10mg/kg, 4 times, every 2h) in the striatum of mice with transgenic PTN overexpression (PTN-Tg) in the brain and in wild type (WT) mice. Amphetamine caused an enhanced loss of striatal dopaminergic terminals, together with a highly significant aggravation of amphetamine-induced increase in the number of GFAP-positive astrocytes, in the striatum of PTN-Tg mice compared to WT mice. Given the known contribution of D1 and D2 dopamine receptors to the neurotoxic effects of amphetamine, we also performed quantitative receptor autoradiography of both receptors in the brains of PTN-Tg and WT mice. D1 and D2 receptors binding in the striatum and other regions of interest was not altered by genotype or treatment. Finally, we found that amphetamine CPP was significantly reduced in PTN-Tg mice. The data demonstrate that PTN overexpression in the brain blocks the conditioning effects of amphetamine and enhances the characteristic striatal dopaminergic denervation caused by this drug. These results indicate for the first time deleterious effects of PTN in vivo by mechanisms that are probably independent of changes in the expression of D1 and D2 dopamine receptors. The data also suggest that PTN-induced neuroinflammation could be involved in the enhanced neurotoxic effects of amphetamine in the striatum of PTN-Tg mice.


Asunto(s)
Anfetamina/farmacología , Proteínas Portadoras/biosíntesis , Estimulantes del Sistema Nervioso Central/farmacología , Cuerpo Estriado/metabolismo , Citocinas/biosíntesis , Neuronas Dopaminérgicas/efectos de los fármacos , Inflamación/metabolismo , Receptores de Dopamina D1/biosíntesis , Receptores de Dopamina D2/biosíntesis , Animales , Astrocitos/efectos de los fármacos , Autorradiografía , Proteínas Portadoras/genética , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Citocinas/genética , Desnervación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
19.
Mediators Inflamm ; 2016: 9894504, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28044069

RESUMEN

Midkine (MK) is a cytokine that modulates amphetamine-induced striatal astrogliosis, suggesting a possible role of MK in neuroinflammation induced by amphetamine. To test this hypothesis, we studied astrogliosis and microglial response induced by amphetamine (10 mg/kg i.p. four times, every 2 h) in different brain areas of MK-/- mice and wild type (WT) mice. We found that amphetamine-induced microgliosis and astrocytosis are enhanced in the striatum of MK-/- mice in a region-specific manner. Surprisingly, LPS-induced astrogliosis in the striatum was blocked in MK-/- mice. Since striatal neuroinflammation induced by amphetamine-type stimulants correlates with the cognitive deficits induced by these drugs, we also tested the long-term effects of periadolescent amphetamine treatment (3 mg/kg i.p. daily for 10 days) in a memory task in MK-/- and WT mice. Significant deficits in the Y-maze test were only observed in amphetamine-pretreated MK-/- mice. The data demonstrate for the first time that MK is a novel modulator of neuroinflammation depending on the inflammatory stimulus and the brain area considered. The data indicate that MK limits amphetamine-induced striatal neuroinflammation. In addition, our data demonstrate that periadolescent amphetamine treatment in mice results in transient disruption of learning and memory processes in absence of endogenous MK.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Cuerpo Estriado/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Anfetaminas/farmacología , Animales , Conducta Animal , Estimulantes del Sistema Nervioso Central/farmacología , Trastornos del Conocimiento/inducido químicamente , Disfunción Cognitiva/inducido químicamente , Cuerpo Estriado/efectos de los fármacos , Gliosis/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inflamación , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Noqueados , Midkina
20.
Neurosci Lett ; 604: 75-9, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26222257

RESUMEN

Pleiotrophin (PTN) is a neurotrophic factor with important functions in addiction and neurodegenerative disorders. Morphine administration induces an increase in the expression of PTN and Midkine (MK), the only other member of this family of cytokines, in brain areas related with the addictive effects of drug of abuse, like the Ventral Tegmental Area or the hippocampus. In spite of previous studies showing that PTN modulates amphetamine and ethanol rewarding effects, and that PTN is involved in morphine-induced analgesia, it was still unknown if the rewarding effects of morphine may be regulated by endogenous PTN. Thus, we aim to study the role of PTN in the reward and physical dependence induced by morphine. We used the Conditioned Place Preference (CPP) paradigm in PTN genetically deficient (PTN-/-) and wild type (WT) mice to assess the rewarding effects of morphine in absence of endogenous PTN. Second, to study if PTN may be involved in morphine physical dependence, naloxone-precipitated withdrawal syndrome was induced in PTN-/- and WT morphine dependent mice. Although the increase in the time spent in the morphine-paired compartment after conditioning tended to be more pronounced in PTN-/- mice, statistical significance was not achieved. The data suggest that PTN does not exert an important role in morphine reward. However, our results clearly indicate that PTN-/- mice develop a more severe withdrawal syndrome than WT mice, characterized as a significant increase in the time standing and in the total incidences of forepaw licking, forepaw tremors, wet dog shake and writhing. The data presented here suggest that PTN is a novel genetic factor that plays a role in morphine withdrawal syndrome.


Asunto(s)
Proteínas Portadoras/metabolismo , Condicionamiento Clásico/efectos de los fármacos , Citocinas/metabolismo , Dependencia de Morfina/metabolismo , Morfina/efectos adversos , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Proteínas Portadoras/genética , Citocinas/genética , Ratones Noqueados , Morfina/farmacología , Dependencia de Morfina/fisiopatología , Dependencia de Morfina/psicología , Naloxona/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Recompensa , Síndrome de Abstinencia a Sustancias/fisiopatología , Síndrome de Abstinencia a Sustancias/psicología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...