Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
J Pharmacol Exp Ther ; 365(3): 652-663, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29632236

RESUMEN

Phytocannabinoids modulate inflammatory responses by regulating the production of cytokines in several experimental models of inflammation. Cannabinoid type-2 (CB2) receptor activation was shown to reduce the production of the monocyte chemotactic protein-2 (MCP-2) chemokine in polyinosinic-polycytidylic acid [poly-(I:C)]-stimulated human keratinocyte (HaCaT) cells, an in vitro model of allergic contact dermatitis (ACD). We investigated if nonpsychotropic cannabinoids, such as cannabidiol (CBD), produced similar effects in this experimental model of ACD. HaCaT cells were stimulated with poly-(I:C), and the release of chemokines and cytokines was measured in the presence of CBD or other phytocannabinoids (such as cannabidiol acid, cannabidivarin, cannabidivarinic acid, cannabichromene, cannabigerol, cannabigerolic acid, cannabigevarin, tetrahydrocannabivarin, and tetrahydrocannabivarinic acid) and antagonists of CB1, CB2, or transient receptor potential vanilloid type-1 (TRPV1) receptors. HaCaT cell viability following phytocannabinoid treatment was also measured. The cellular levels of endocannabinoids [anandamide (AEA), 2-arachidonoylglycerol] and related molecules (palmitoylethanolamide, oleoylethanolamide) were quantified in poly-(I:C)-stimulated HaCaT cells treated with CBD. We show that in poly-(I:C)-stimulated HaCaT cells, CBD elevates the levels of AEA and dose-dependently inhibits poly-(I:C)-induced release of MCP-2, interleukin-6 (IL-6), IL-8, and tumor necrosis factor-α in a manner reversed by CB2 and TRPV1 antagonists 6-iodopravadoline (AM630) and 5'-iodio-resiniferatoxin (I-RTX), respectively, with no cytotoxic effect. This is the first demonstration of the anti-inflammatory properties of CBD in an experimental model of ACD.


Asunto(s)
Antiinflamatorios/farmacología , Cannabidiol/farmacología , Dermatitis Alérgica por Contacto/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Ácidos Araquidónicos/metabolismo , Cannabidiol/uso terapéutico , Línea Celular , Quimiocina CCL8/metabolismo , Dermatitis Alérgica por Contacto/metabolismo , Endocannabinoides/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Alcamidas Poliinsaturadas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
2.
Eur J Pharmacol ; 791: 669-674, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27720681

RESUMEN

In mice, 2,4-dinitrofluorobenzene (DNFB) induces contact allergic dermatitis (CAD), which, in a late phase, is characterized by mast cell (MC) infiltration and angiogenesis. Palmitoylethanolamide (PEA), an endogenous anti-inflammatory molecule, acts by down-modulating MCs following activation of the cannabinoid CB2 receptor and peroxisome proliferator-activated receptor-α (PPAR-α). We have previously reported the anti-inflammatory effect of PEA in the early stage of CAD. Here, we examined whether PEA reduces the features of the late stage of CAD including MC activation, angiogenesis and itching. After sensitization to DNFB, female C57BL/6J mice underwent to three DNFB challenges at days 5, 12 and 19 and treatments were given at each challenge and for two more days. CAD was expressed as Δ increase in ear thickness between challenged and un-challenged mice. PEA (5mg/kg/i.p.) reduced: i) the DNFB-induced Δ increase; ii) the number of MCs per tissue area; iii) the expression of VEGF and its receptor Flk-1. These effects were reversed by co-administration of AM630 (1mg/kg/i.p.), a CB2 antagonist, but not GW6471 (1mg/kg/i.p.), a PPAR-α antagonist. Finally, PEA reduced the number of ear scratchings 48h after DNFB challenge and this effect was reversed by both CB2 and PPAR-α antagonists, suggesting the involvement of both receptors. PEA, by reducing the features of late stage CAD in mice, may be beneficial in this pathological condition.


Asunto(s)
Dermatitis Alérgica por Contacto/complicaciones , Dermatitis Alérgica por Contacto/tratamiento farmacológico , Etanolaminas/farmacología , Ácidos Palmíticos/farmacología , Prurito/complicaciones , Amidas , Animales , Recuento de Células , Dermatitis Alérgica por Contacto/inmunología , Dermatitis Alérgica por Contacto/metabolismo , Dinitrofluorobenceno/efectos adversos , Modelos Animales de Enfermedad , Endocannabinoides/metabolismo , Etanolaminas/uso terapéutico , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/complicaciones , Mastocitos/citología , Mastocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ácidos Palmíticos/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Biochem Pharmacol ; 112: 72-81, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27188793

RESUMEN

Extracellular adenosine formation from ATP is controlled by ecto-nucleoside triphosphate diphosphohydrolase (E-NTPDase/CD39) and ecto-5'-nucleotidase (e-5NT/CD73); the latter converts AMP to adenosine and inorganic phosphate, representing the rate limiting step controlling the ratio between extracellular ATP and adenosine. Evidence that cellular expression and activity of CD39 and CD73 may be subject to changes under pathophysiological conditions has identified this pathway as an endogenous modulator in several diseases and was shown to be involved in the molecular mechanism of drugs, such as methotrexate, salicylates , interferon-ß. We evaluated whether CD73/adenosine/A2A signalling pathway is involved in nimesulide anti-inflammatory effect, in vivo and in vitro. We found that the adenosine A2A agonist, 4-[2-[[6-amino-9-(N-ethyl-ß-d-ribofuranuronamidosyl)-9H-purin-2-yl]amino]ethyl]benzenepropanoic acid hydrochloride (CGS21680, 2mg/kg ip.), inhibited carrageenan-induced rat paw oedema and the effect was reversed by co-administration of the A2A antagonist -(2-[7-amino-2-[2-furyl][1,2,4]triazolo[2,3-a][1,3,5]triazin-5-yl-amino]ethyl)phenol (ZM241385; 3mg/kg i.p.). Nimesulide (5mg/kg i.p.) anti-inflammatory effect was inhibited by pre-treatment with ZM241385 (3mg/kg i.p.) and by local administration of the CD73 inhibitor, adenosine 5'-(α,ß-methylene)diphosphate (APCP; 400µg/paw). Furthermore, we found increased activity of 5'-nucleotidase/CD73 in paws and plasma of nimesulide treated rats, 4h following oedema induction. In vitro, the inhibitory effect of nimesulide on nitrite and prostaglandin E2 production by lipopolysaccharide-activated J774 cell line was reversed by ZM241385 and APCP. Furthermore, nimesulide increased CD73 activity in J774 macrophages while it did not inhibit nitrite accumulation by lipopolysaccharide-activated SiRNA CD73 silenced J774 macrophages. Our data demonstrate that the anti-inflammatory effect of nimesulide in part is mediated by CD73-derived adenosine acting on A2A receptors.


Asunto(s)
Adenosina/metabolismo , Antiinflamatorios/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , 5'-Nucleotidasa/antagonistas & inhibidores , 5'-Nucleotidasa/sangre , 5'-Nucleotidasa/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Línea Celular , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Dinoprostona/sangre , Edema/tratamiento farmacológico , Edema/inmunología , Edema/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratas Wistar , Receptor de Adenosina A2A/metabolismo , Sulfonamidas/administración & dosificación
4.
PLoS One ; 11(5): e0156198, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27219328

RESUMEN

BACKGROUND AND AIM: Angiogenesis is emerging as a pivotal process in chronic inflammatory pathologies, promoting immune infiltration and prompting carcinogenesis. Ulcerative Colitis (UC) and Crohn's Disease (CD) represent paradigmatic examples of intestinal chronic inflammatory conditions in which the process of neovascularization correlates with the severity and progression of the diseases. Molecules able to target the angiogenesis have thus the potential to synergistically affect the disease course. Beyond its anti-inflammatory effect, palmitoylethanolamide (PEA) is able to reduce angiogenesis in several chronic inflammatory conditions, but no data about its anti-angiogenic activity in colitis have been produced, yet. METHODS: The effects of PEA on inflammation-associated angiogenesis in mice with dextran sulphate sodium (DSS)-induced colitis and in patients with UC were assessed. The release of Vascular Endothelial Growth Factor (VEGF), the hemoglobin tissue content, the expression of CD31 and of phosphatidylinositol 3-kinase/Akt/mammalian-target-of-rapamycin (mTOR) signaling axis were all evaluated in the presence of different concentrations of PEA and concomitant administration of PPAR-α and -γ antagonists. RESULTS: Our results demonstrated that PEA, in a selective peroxisome proliferator activated receptor (PPAR)-α dependent mechanism, inhibits colitis-associated angiogenesis, decreasing VEGF release and new vessels formation. Furthermore, we demonstrated that the mTOR/Akt axis regulates, at least partly, the angiogenic process in IBD and that PEA directly affects this pathway. CONCLUSIONS: Our results suggest that PEA may improve inflammation-driven angiogenesis in colonic mucosa, thus reducing the mucosal damage and potentially affecting disease progression and the shift towards the carcinogenesis.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Colitis/tratamiento farmacológico , Sulfato de Dextran/efectos adversos , Etanolaminas/administración & dosificación , PPAR alfa/metabolismo , Ácidos Palmíticos/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto , Amidas , Animales , Células Cultivadas , Colitis/inducido químicamente , Colitis/metabolismo , Colitis Ulcerosa/metabolismo , Modelos Animales de Enfermedad , Etanolaminas/farmacología , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Ácidos Palmíticos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
5.
Phytother Res ; 30(6): 963-70, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26929026

RESUMEN

Palmitoylethanolamide (PEA) is a nutraceutical compound that has been demonstrated to improve intestinal inflammation. We aimed at evaluating its antiproliferative and antiangiogenic effects in human colon adenocarcinoma Caco-2 cell line. Caco-2 cells were treated with increasing concentrations of PEA (0.001, 0.01 and 0.1 µM) in the presence of peroxisome proliferator-activated receptor-a (PPAR-α) or PPAR-γ antagonists. Cell proliferation was evaluated by performing a MTT assay. Vascular endothelial growth factor (VEGF) release was estimated by ELISA, while the expression of VEGF receptor and the activation of the Akt/mammalian target of rapamycin (mTOR) pathway were evaluated by western blot analysis. PEA caused a significant and concentration-dependent decrease of Caco-2 cell proliferation at 48 h. PEA administration significantly reduced in a concentration-dependent manner VEGF secretion and VEGF receptor expression. Inhibition of Akt phosphorylation and a downstream decrease of phospho-mTOR and of p-p70S6K were observed as compared with untreated cells. PPAR-α, but not PPAR-γ antagonist, reverted all effects of PEA. PEA is able to decrease cell proliferation and angiogenesis. The antiangiogenic effect of PEA depends on the specific inhibition of the AkT/mTOR axis, through the activation of PPAR-α pathway. If supported by in vivo models, our data pave the way to PEA co-administration to the current chemotherapeutic regimens for colon carcinoma. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Etanolaminas/química , PPAR alfa/metabolismo , Ácidos Palmíticos/química , Factor A de Crecimiento Endotelial Vascular/metabolismo , Amidas , Animales , Células CACO-2 , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo/efectos de los fármacos , Etanolaminas/uso terapéutico , Humanos , Neovascularización Patológica , Ácidos Palmíticos/uso terapéutico , Transducción de Señal
7.
Biochem Pharmacol ; 104: 83-94, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26776306

RESUMEN

Nuclear Factor-κB (NF-κB) is a transcription factor regulating several genes involved in important physiological and pathological processes. NF-κB has been found constitutively activated in many inflammatory/immune diseases. In addition, a positive correlation between persistent activation of NF-κB and tumor promotion has been demonstrated. Since the IKK (IκB kinase) activation is an indispensable component of all pro-inflammatory signaling pathways leading to NF-κB activation, considerable efforts have been done in order to develop novel anti-inflammatory therapeutics targeting IKK. Association of the IKK complex relies on critical interactions between the C-terminus NBD (NEMO binding domain) of the catalytic subunits IKKα and IKKß, and the regulatory subunit NEMO (NF-κB Essential Modulator). Thus, this IKK/NEMO interacting region provides an attractive target to prevent the IKK complex formation and NF-κB activation. In this regard, we have identified non-peptide small molecule disruptors of IKKß/NEMO complex through a structure-based virtual screening (SBVS) of the NCI chemical library. Phenothiazine 22 and its close analogues (22.2, 22.4 and 22.10) were able to reduce nitrite production and iNOS mRNA expression in J774 murine macrophages stimulated with LPS for 24h. These effects were associated with a reduced NF-κB/DNA binding activity as well as a decreased expression of phosphorylated IKKß, IκBα and NF-κB/p65 in these cells. These observations suggest that compound 22 and its three structural analogues by inhibiting IKKß/NEMO association mediate the blockage of NF-κB signaling pathway and may prove effective in treatment of diseases in which the IKK/NF-κB pathway is dysregulated.


Asunto(s)
Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Sitios de Unión , Línea Celular , Supervivencia Celular/efectos de los fármacos , Descubrimiento de Drogas , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Modelos Moleculares , Estructura Molecular , Unión Proteica , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/química
8.
Pain ; 157(1): 80-91, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25974242

RESUMEN

The effects of ultramicronized palmitoylethanolamide were evaluated on pain behaviours and markers of mast cell (MC) activity in a rat model of endometriosis plus ureteral calculosis (ENDO+STONE)-induced viscerovisceral hyperalgesia (VVH). Female Sprague-Dawley rats that underwent surgical induction of endometriosis were randomly assigned to receive active (ultramicronized palmitoylethanolamide 10 mg·kg(-1)·d(-1), orally) or placebo treatment for 25 days. At day 21, they underwent ureteral stone formation and were video-recorded till day 25 to evaluate ureteral and uterine pain behaviours. At autopsy (day 25), ureteral condition and number and diameter of endometrial cysts were evaluated. The following were then measured: number and percentage of degranulating MCs, number of vessels, chymase, nerve growth factor (NGF), vascular endothelial growth factor (VEGF), and Flk-1 (VEGF receptor) in cysts, and NGF in dorsal root ganglia (DRG). Ultramicronized palmitoylethanolamide-treated vs placebo-treated rats showed significantly lower number, duration and complexity of ureteral crises, shorter duration of uterine pain, and smaller cyst diameter (0.0001 < P < 0.004); a significantly higher percentage of expelled stones (P < 0.0001); significantly lower MC number (P < 0.01), vessel number (P < 0.01), chymase (P < 0.05), NGF (P < 0.05), VEGF (P < 0.01), and Flk-1 (P < 0.01) expression in cysts and NGF expression in DRG (P < 0.01). In all animals, the global duration of ureteral crises correlated linearly and directly with cyst diameter, MC number and chymase in cysts, and NGF in cysts and DRG (0.02 < P < 0.0002). Ultramicronized palmitoylethanolamide significantly reduces VVH from ENDO+STONE, probably by modulating MC expression/activity in cysts, thus reducing central sensitization due to noxious signals from endometriotic lesions. The results suggest potential utility of the compound for VVH in clinics.


Asunto(s)
Endometriosis/complicaciones , Etanolaminas/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Mastocitos/efectos de los fármacos , Ácidos Palmíticos/uso terapéutico , Cálculos Ureterales/complicaciones , Amidas , Animales , Quimasas/metabolismo , Modelos Animales de Enfermedad , Endometriosis/metabolismo , Etanolaminas/farmacología , Femenino , Hiperalgesia/complicaciones , Hiperalgesia/metabolismo , Mastocitos/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Ácidos Palmíticos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Cálculos Ureterales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Biomed Res Int ; 2015: 508342, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26295040

RESUMEN

Among the different signaling molecules released during reactive gliosis occurring in Alzheimer's disease (AD), the astrocyte-derived S100B protein plays a key role in neuroinflammation, one of the hallmarks of the disease. The use of pharmacological tools targeting S100B may be crucial to embank its effects and some of the pathological features of AD. The antiprotozoal drug pentamidine is a good candidate since it directly blocks S100B activity by inhibiting its interaction with the tumor suppressor p53. We used a mouse model of amyloid beta- (Aß-) induced AD, which is characterized by reactive gliosis and neuroinflammation in the brain, and we evaluated the effect of pentamidine on the main S100B-mediated events. Pentamidine caused the reduction of glial fibrillary acidic protein, S100B, and RAGE protein expression, which are signs of reactive gliosis, and induced p53 expression in astrocytes. Pentamidine also reduced the expression of proinflammatory mediators and markers, thus reducing neuroinflammation in AD brain. In parallel, we observed a significant neuroprotection exerted by pentamidine on CA1 pyramidal neurons. We demonstrated that pentamidine inhibits Aß-induced gliosis and neuroinflammation in an animal model of AD, thus playing a role in slowing down the course of the disease.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/tratamiento farmacológico , Gliosis/complicaciones , Gliosis/tratamiento farmacológico , Neuronas/patología , Pentamidina/uso terapéutico , Subunidad beta de la Proteína de Unión al Calcio S100/antagonistas & inhibidores , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Densitometría , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Ensayo de Cambio de Movilidad Electroforética , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/patología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Humanos , Inmunohistoquímica , Inflamación/complicaciones , Inflamación/patología , Inyecciones , Interleucina-1beta/metabolismo , Malondialdehído/metabolismo , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nitritos/metabolismo , Pentamidina/farmacología , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
10.
Brain Res ; 1610: 89-97, 2015 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-25813826

RESUMEN

It has already been reported that cannabinoids are neuroprotective agents against excitotoxicity in vitro and increase after acute brain damage in vivo. This background prompted us to study the localization and expression of the calcium -binding protein calretinin in a condition similar to Alzheimer disease and its possible relationship with cannabinoids and their supposed protective role. We carried out quantitative analysis of the transient changes in calretinin expression shown by hybridochemistry within neuronal cell populations in the hippocampus of a beta amyloid-treated rat model of Alzheimer's disease and their correlation with endocannabinoid increase. Calretinin expression increases throughout the first week after cortical amyloid-beta peptide injection, and then decreases towards normal levels in the rat hippocampus during the following weeks, indicating that decreased calretinin gene expression may be associated with either increase of endocannabinoids or VDM11-induced accumulation of endocannabinoids. In contrast, SR1, an antagonist, which limits the cannabinoid effect by selective binding to the cannabinoid receptor CB1, up-regulates calretinin expression with respect to non-treated rats. This could mean that the SR1 endocannabinoid-blocking action through CB1 receptors, that are normally stimulated by endocannabinoids to inhibit calcium increase, might cause a higher calretinin expression. This would allow us to speculate on a possible reverse relationship between endocannabinoid and calretinin levels in the hippocampal calcium-homeostasis balance.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Calbindina 2/metabolismo , Endocannabinoides/metabolismo , Hipocampo/metabolismo , Fragmentos de Péptidos/toxicidad , Enfermedad de Alzheimer/metabolismo , Animales , Modelos Animales de Enfermedad , Expresión Génica , Hipocampo/efectos de los fármacos , Masculino , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas Wistar , Receptor Cannabinoide CB1/metabolismo , Factores de Tiempo
11.
CNS Neurol Disord Drug Targets ; 14(7): 828-37, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25801844

RESUMEN

Aß-induced astrogliosis can worsen the eziopathogenesis of Alzheimer disease (AD) by the release of proinflammatory and pro-oxidant mediators. Activated glial cells may release also pro-angiogenic molecules. The role of angiogenesis in AD is still controversial: although angiogenesis brings oxygen and nutrients to injured tissue, it may also exacerbate reactive gliosis. Moreover, by altering blood-brain barrier permeability pro-angiogenic mediators promote passage of inflammatory/immune-competent cells into the brain, thereby exacerbating gliosis. The release of proangiogenic factors during astrogliosis may thus be a key-step in controlling AD progression. The endogenous fatty acid amide, palmitoylethanolamide (PEA), is a pleiotropic mediator exerting anti-inflammatory, antinociceptive and antiangiogenic effects in several in vitro and in vivo models of chronic-degenerative disease. In this study, we investigated the effects of PEA in AD angiogenesis and neuroinflammation by using conditioned medium from untreated and Aß-treated C6 rat astroglioma cells and HUVEC human endothelial cells. PEA (10-8-10-6 M) concentration-dependently reduced expression of pro-inflammatory and pro-angiogenic markers in Aß (1 µg/mL)-stimulated C6 cells. Moreover, culture medium from PEA-treated C6 cells reduced HUVEC cell proliferation as compared to cells treated with conditioned medium from Aß-treated C6 cells. Immunocytochemical analysis revealed that PEA treatment inhibited nuclear levels of mitogen-activated protein kinase 1 (the main pro-angiogenic pathway) and cytoplasmic vascular endothelial growth factor in HUVEC cells receiving C6 conditioned medium. Finally, the peroxisome proliferator-activated receptor alpha inhibitor GW6471, added to Aß-treated C6 cells blocked all PEA effects in this model, suggesting that PEA acts through a proliferator-activated receptor alpha-dependent mechanism on astroglial cells. Collectively, these data support the potential therapeutic utility of PEA in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Astrocitos/efectos de los fármacos , Etanolaminas/farmacología , Gliosis/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Ácidos Palmíticos/farmacología , Fragmentos de Péptidos/metabolismo , Adenosina Trifosfato/metabolismo , Amidas , Animales , Astrocitos/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Relación Dosis-Respuesta a Droga , Gliosis/metabolismo , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Oxazoles/farmacología , PPAR alfa/antagonistas & inhibidores , PPAR alfa/metabolismo , Ratas , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Tirosina/análogos & derivados , Tirosina/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Eur J Pharmacol ; 725: 64-9, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24440533

RESUMEN

Chronic inflammation, a condition frequently associated with several pathologies, is characterized by angiogenic and fibrogenic responses that may account for the development of granulomatous tissue. We previously demonstrated that the chymase, rat mast cell protease-5 (rMCP-5), exhibits pro-inflammatory and pro-angiogenic properties in a model of chronic inflammation sustained by mast cells (MCs), granuloma induced by the subcutaneous carrageenan-soaked sponge implant in rat. In this study, we investigated the effects of palmitoylethanolamide (PEA), an anti-inflammatory and analgesic endogenous compound, on rMCP-5 mRNA expression and Microphtalmia-associated Transcription Factor (MITF) activation in the same model of chronic inflammation. The levels of rMCP-5 mRNA were detected using semi-quantitative RT-PCR; the protein expression of chymase and extracellular signal-regulated kinases (ERK) were analyzed by western blot; MITF/DNA binding activity and MITF phosphorylation were assessed by electrophoretic mobility shift assay (EMSA) and immunoprecipitation, respectively. The administration of PEA (200, 400 and 800 µg/ml) significantly decreased rMCP-5 mRNA and chymase protein expression induced by λ-carrageenan. These effects were associated with a significant decrease of MITF/DNA binding activity and phosphorylated MITF as well as phosphorylated ERK levels. In conclusion, our results, showing the ability of PEA to inhibit MITF activation and chymase expression in granulomatous tissue, may yield new insights into the understanding of the signaling pathways leading to MITF activation controlled by PEA.


Asunto(s)
Quimasas/genética , Endocannabinoides/farmacología , Etanolaminas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Granuloma/genética , Granuloma/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Ácidos Palmíticos/farmacología , Amidas , Animales , Carragenina/efectos adversos , Enfermedad Crónica , ADN/metabolismo , Endocannabinoides/uso terapéutico , Etanolaminas/uso terapéutico , Granuloma/tratamiento farmacológico , Granuloma/patología , Masculino , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Ácidos Palmíticos/uso terapéutico , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar , Transcripción Genética/efectos de los fármacos
13.
Phytother Res ; 27(5): 633-6, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22815234

RESUMEN

This minireview highlights the importance of cannabidiol (CBD) as a promising drug for the therapy of inflammatory bowel diseases (IBD). Actual pharmacological treatments for IBD should be enlarged toward the search for low-toxicityand low-cost drugs that may be given alone or in combination with the conventional anti-IBD drugs to increase their efficacy in the therapy of relapsing forms of colitis. In the past, Cannabis preparations have been considered new promising pharmacological tools in view of their anti-inflammatory role in IBD as well as other gut disturbances. However, their use in the clinical therapy has been strongly limited by their psychotropic effects. CBD is a very promising compound since it shares the typical cannabinoid beneficial effects on gut lacking any psychotropic effects. For years, its activity has been enigmatic for gastroenterologists and pharmacologists, but now it is evident that this compound may interact at extra-cannabinoid system receptor sites, such as peroxisome proliferator-activated receptor-gamma. This strategic interaction makes CBD as a potential candidate for the development of a new class of anti-IBD drugs.


Asunto(s)
Cannabidiol/uso terapéutico , Fármacos Gastrointestinales/uso terapéutico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Humanos , Inflamación/tratamiento farmacológico , Fitoterapia , Extractos Vegetales/uso terapéutico
14.
PLoS One ; 7(10): e47464, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23077623

RESUMEN

Bindarit, a selective inhibitor of monocyte chemotactic proteins (MCPs) synthesis, reduces neointimal formation in animal models of vascular injury and recently has been shown to inhibit in-stent late loss in a placebo-controlled phase II clinical trial. However, the mechanisms underlying the efficacy of bindarit in controlling neointimal formation/restenosis have not been fully elucidated. Therefore, we investigated the effect of bindarit on human coronary smooth muscle cells activation, drawing attention to the phenotypic modulation process, focusing on contractile proteins expression as well as proliferation and migration. The expression of contractile proteins was evaluated by western blot analysis on cultured human coronary smooth muscle cells stimulated with TNF-α (30 ng/mL) or fetal bovine serum (5%). Bindarit (100-300 µM) reduced the embryonic form of smooth muscle myosin heavy chain while increased smooth muscle α-actin and calponin in both TNF-α- and fetal bovine serum-stimulated cells. These effects were associated with the inhibition of human coronary smooth muscle cell proliferation/migration and both MCP-1 and MCP-3 production. The effect of bindarit on smooth muscle cells phenotypic switching was confirmed in vivo in the rat balloon angioplasty model. Bindarit (200 mg/Kg/day) significantly reduced the expression of the embryonic form of smooth muscle myosin heavy chain, and increased smooth muscle α-actin and calponin in the rat carodid arteries subjected to endothelial denudation. Our results demonstrate that bindarit induces the differentiated state of human coronary smooth muscle cells, suggesting a novel underlying mechanisms by which this drug inhibits neointimal formation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Indazoles/administración & dosificación , Proteínas Quimioatrayentes de Monocitos/metabolismo , Propionatos/administración & dosificación , Actinas/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Movimiento Celular/fisiología , Vasos Coronarios/citología , Vasos Coronarios/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Microfilamentos/metabolismo , Proteínas Quimioatrayentes de Monocitos/antagonistas & inhibidores , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Cadenas Pesadas de Miosina/metabolismo , Neointima/patología , Ratas , Calponinas
15.
PLoS One ; 6(12): e28159, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22163000

RESUMEN

Enteric glial cells (EGC) actively mediate acute and chronic inflammation in the gut; EGC proliferate and release neurotrophins, growth factors, and pro-inflammatory cytokines which, in turn, may amplify the immune response, representing a very important link between the nervous and immune systems in the intestine. Cannabidiol (CBD) is an interesting compound because of its ability to control reactive gliosis in the CNS, without any unwanted psychotropic effects. Therefore the rationale of our study was to investigate the effect of CBD on intestinal biopsies from patients with ulcerative colitis (UC) and from intestinal segments of mice with LPS-induced intestinal inflammation. CBD markedly counteracted reactive enteric gliosis in LPS-mice trough the massive reduction of astroglial signalling neurotrophin S100B. Histological, biochemical and immunohistochemical data demonstrated that S100B decrease was associated with a considerable decrease in mast cell and macrophages in the intestine of LPS-treated mice after CBD treatment. Moreover the treatment of LPS-mice with CBD reduced TNF-α expression and the presence of cleaved caspase-3. Similar results were obtained in ex vivo cultured human derived colonic biopsies. In biopsies of UC patients, both during active inflammation and in remission stimulated with LPS+INF-γ, an increased glial cell activation and intestinal damage were evidenced. CBD reduced the expression of S100B and iNOS proteins in the human biopsies confirming its well documented effect in septic mice. The activity of CBD is, at least partly, mediated via the selective PPAR-gamma receptor pathway. CBD targets enteric reactive gliosis, counteracts the inflammatory environment induced by LPS in mice and in human colonic cultures derived from UC patients. These actions lead to a reduction of intestinal damage mediated by PPARgamma receptor pathway. Our results therefore indicate that CBD indeed unravels a new therapeutic strategy to treat inflammatory bowel diseases.


Asunto(s)
Cannabidiol/farmacología , Inflamación/metabolismo , Intestinos/efectos de los fármacos , Animales , Biopsia , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/patología , Colon/inmunología , Colon/metabolismo , Humanos , Sistema Inmunológico , Inflamación/tratamiento farmacológico , Interferón gamma/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos/citología , Masculino , Mastocitos/citología , Ratones , Sistema Nervioso/metabolismo , Nitritos/metabolismo , Sepsis
16.
PLoS One ; 6(12): e28668, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22163051

RESUMEN

Peroxisome proliferator-activated receptor-γ (PPARγ) has been reported to be involved in the etiology of pathological features of Alzheimer's disease (AD). Cannabidiol (CBD), a Cannabis derivative devoid of psychomimetic effects, has attracted much attention because of its promising neuroprotective properties in rat AD models, even though the mechanism responsible for such actions remains unknown. This study was aimed at exploring whether CBD effects could be subordinate to its activity at PPARγ, which has been recently indicated as its putative binding site. CBD actions on ß-amyloid-induced neurotoxicity in rat AD models, either in presence or absence of PPAR antagonists were investigated. Results showed that the blockade of PPARγ was able to significantly blunt CBD effects on reactive gliosis and subsequently on neuronal damage. Moreover, due to its interaction at PPARγ, CBD was observed to stimulate hippocampal neurogenesis. All these findings report the inescapable role of this receptor in mediating CBD actions, here reported.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Cannabidiol/farmacología , Hipocampo/metabolismo , Neurogénesis , PPAR gamma/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Sitios de Unión , Encéfalo/metabolismo , Inflamación/metabolismo , Masculino , Neuronas/metabolismo , Neuronas/patología , Óxido Nítrico/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
17.
J Cell Mol Med ; 15(12): 2664-74, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21255263

RESUMEN

Emerging evidence indicates that astrogliosis is involved in the pathogenesis of neurodegenerative disorders. Our previous findings suggested cannabinoids and Autacoid Local Injury Antagonism Amides (ALIAmides) attenuate glial response in models of neurodegeneration. The present study was aimed at exploring palmitoylethanolamide (PEA) ability to mitigate ß-amyloid (Aß)-induced astrogliosis. Experiments were carried out to investigate PEA's (10(-7) M) effects upon the expression and release of pro-inflammatory molecules in rat primary astrocytes activated by soluble Aß(1-42) (1 µg/ml) as well as to identify mechanisms responsible for such actions. The effects of Aß and exogenous PEA on the astrocyte levels of the endocannabinoidsand of endogenous ALIAmides were also studied. The peroxisome proliferator-activated receptor (PPAR)-α (MK886, 3 µM) or PPAR-γ (GW9662, 9 nM) antagonists were co-administered with PEA. Aß elevated endogenous PEA and d5-2-arachidonoylglycerol (2-AG) levels. Exogenous PEA blunted the Aß-induced expression of pro-inflammatory molecules. This effect was reduced by PPAR-α antagonist. Moreover, this ALIAmide, like Aß, increased 2-AG levels. These results indicate that PEA exhibits anti-inflammatory properties able to counteract Aß-induced astrogliosis, and suggest novel treatment for neuroinflammatory/ neurodegenerative processes.


Asunto(s)
Péptidos beta-Amiloides/efectos adversos , Antiinflamatorios no Esteroideos/farmacología , Astrocitos/efectos de los fármacos , Gliosis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Ácidos Palmíticos/farmacología , Fragmentos de Péptidos/efectos adversos , Amidas , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/metabolismo , Western Blotting , Ensayo de Cambio de Movilidad Electroforética , Endocannabinoides , Etanolaminas , Técnica del Anticuerpo Fluorescente , Gliosis/etiología , Humanos , Inflamación/etiología , PPAR alfa/antagonistas & inhibidores , PPAR alfa/metabolismo , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
Mol Pain ; 7: 3, 2011 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-21219627

RESUMEN

The aim of this study was to obtain evidences of a possible analgesic role for palmitoylethanolamide (PEA) in chronic granulomatous inflammation sustained by mast cell (MC) activation in rats at 96 hours. PEA (200-400-800 µg/mL), locally administered at time 0, reduced in a concentration-dependent manner the expression and release of NGF in comparison with saline-treated controls. PEA prevented nerve formation and sprouting, as shown by histological analysis, reduced mechanical allodynia, evaluated by Von Frey filaments, and inhibited dorsal root ganglia activation. These results were supported by the evidence that MCs in granuloma were mainly degranulated and closely localized near nerve fibres and PEA significantly reduced MC degranulation and nerves fibre formation. These findings are the first evidence that PEA, by the modulation of MC activation, controls pain perception in an animal model of chronic inflammation, suggesting its potential use for the treatment of all those painful conditions in which MC activation is an initial key step.


Asunto(s)
Granuloma/complicaciones , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Mastocitos/patología , Ácidos Palmíticos/uso terapéutico , Amidas , Animales , Carragenina , Endocannabinoides , Etanolaminas , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Granuloma/patología , Hiperalgesia/patología , Masculino , Mastocitos/efectos de los fármacos , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/patología , Factores de Crecimiento Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Ácidos Palmíticos/administración & dosificación , Ácidos Palmíticos/farmacología , Ratas , Ratas Wistar , Factores de Tiempo
19.
CNS Neurol Disord Drug Targets ; 9(6): 807-12, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20874699

RESUMEN

Recently, a pivotal role for neuroinflammation in the pathogenesis of several neurodegenerative diseases has been recognized. Once activated, glial cells produce pathological amounts of neurotoxic substances driving neurodegeneration into chronic progression through a self-propagating cycle. Nevertheless, mounting evidence suggests that also angiogenesis may importantly contribute to neurodegeneration, since activated glial cells may release also pro-angiogenic factors. A deregulation of the balance between pro- and anti-angiogenic mediators has been reported in in vivo and in vitro models of neuroinflammation. Indeed, in Alzheimer's disease brain, a significant increase in the expression of pro-angiogenic growth factors, such as vascular endothelial growth factor, was found strictly co-localized with senile plaques. In addition, converging results indicate that thalidomide and its derivatives, having newly discovered anti-inflammatory and anti-angiogenic properties, are useful in the prevention of several hallmarks of neurodegeneration occurring in experimental models of Parkinson's and Alzheimer's diseases. The present review primarily discusses about the possible roles, still under debate, of angiogenesis in neurodegeneration, and focuses on the identification of new possible anti-angiogenic compounds that could open new horizons in the treatment of neurodegenerative diseases where angiogenesis is detrimental.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Animales , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Enfermedades Neurodegenerativas/fisiopatología
20.
Endocrinology ; 151(3): 921-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20133454

RESUMEN

The endocannabinoid system plays protective roles against the growth and the spreading of several types of carcinomas. Because estrogens regulate this system both in physiological states and cancer, in this paper we evaluated its involvement in endometrial carcinoma, a well-known estrogen-dependent tumor. To test whether the endocannabinoid system is expressed in endometrial cancer, tissue samples were collected both from 18 patients undergoing surgical treatment for endometrial adenocarcinoma and 16 healthy age-matched controls, and treated for Western blot and immunohistochemical analysis. Moreover, tissues were dounce homogenized and submitted to endocannabinoid measurement by liquid chromatography-mass spectrometry. To evaluate the physiological role of the endocannabinoid system, a human endometrial cancer cell-line (AN3CA) was used and transiently transfected with a plasmid containing the cDNA for the endocannabinoid receptor CB(2). Cells were incubated for 48 h with an agonist (JWH133) (10 mum) or antagonist (SR144528) (1 mum) of CB(2) 24 h after transfection, and cell proliferation was measured by the 3-[4,5-dimethyltiazol-2yl]-2,5 diphenyltetrazolium bromide formazan assay. In human endometrial carcinoma biopsies the expression of CB(2) receptor and the levels of its ligand, 2-arachidonoylglycerol increased, whereas monoacylglycerol lipase, an enzyme responsible for 2-arachidonoylglycerol degradation, was down-regulated. Immunohistochemical analysis revealed that CB(2) was overexpressed only in malignant endometrial cells. CB(2)-overexpressing AN3CA cells showed a significant reduction in cell vitality compared with parental AN3CA cells: incubation with the selective CB(2) antagonist SR144128 restored the viability of CB(2)-overexpressing cells to that of untransfected cells. In conclusion, the endocannabinoid system seems to play an important role in human endometrial carcinoma, and modulation of CB(2) activity/expression may account for a tumor-suppressive effect.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Carcinoma/metabolismo , Neoplasias Endometriales/metabolismo , Glicéridos/metabolismo , Receptor Cannabinoide CB2/metabolismo , Adulto , Biopsia , Carcinoma/patología , Línea Celular Tumoral , Regulación hacia Abajo , Endocannabinoides , Neoplasias Endometriales/patología , Endometrio/patología , Femenino , Humanos , Persona de Mediana Edad , Monoacilglicerol Lipasas/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA