Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 791: 669-674, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27720681

ABSTRACT

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.


Subject(s)
Dermatitis, Allergic Contact/complications , Dermatitis, Allergic Contact/drug therapy , Ethanolamines/pharmacology , Palmitic Acids/pharmacology , Pruritus/complications , Amides , Animals , Cell Count , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/metabolism , Dinitrofluorobenzene/adverse effects , Disease Models, Animal , Endocannabinoids/metabolism , Ethanolamines/therapeutic use , Female , Gene Expression Regulation/drug effects , Inflammation/complications , Mast Cells/cytology , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Palmitic Acids/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
3.
Pain ; 157(1): 80-91, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25974242

ABSTRACT

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.


Subject(s)
Endometriosis/complications , Ethanolamines/therapeutic use , Hyperalgesia/drug therapy , Mast Cells/drug effects , Palmitic Acids/therapeutic use , Ureteral Calculi/complications , Amides , Animals , Chymases/metabolism , Disease Models, Animal , Endometriosis/metabolism , Ethanolamines/pharmacology , Female , Hyperalgesia/complications , Hyperalgesia/metabolism , Mast Cells/metabolism , Nerve Growth Factor/metabolism , Palmitic Acids/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Vascular Endothelial Growth Factor/metabolism , Ureteral Calculi/metabolism , Vascular Endothelial Growth Factor A/metabolism
4.
CNS Neurol Disord Drug Targets ; 14(7): 828-37, 2015.
Article in English | MEDLINE | ID: mdl-25801844

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Astrocytes/drug effects , Ethanolamines/pharmacology , Gliosis/drug therapy , Neuroprotective Agents/pharmacology , Palmitic Acids/pharmacology , Peptide Fragments/metabolism , Adenosine Triphosphate/metabolism , Amides , Animals , Astrocytes/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Gliosis/metabolism , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxazoles/pharmacology , PPAR alpha/antagonists & inhibitors , PPAR alpha/metabolism , Rats , S100 Calcium Binding Protein beta Subunit/metabolism , Tyrosine/analogs & derivatives , Tyrosine/pharmacology , Vascular Endothelial Growth Factor A/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Eur J Pharmacol ; 725: 64-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24440533

ABSTRACT

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.


Subject(s)
Chymases/genetics , Endocannabinoids/pharmacology , Ethanolamines/pharmacology , Gene Expression Regulation/drug effects , Granuloma/genetics , Granuloma/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Palmitic Acids/pharmacology , Amides , Animals , Carrageenan/adverse effects , Chronic Disease , DNA/metabolism , Endocannabinoids/therapeutic use , Ethanolamines/therapeutic use , Granuloma/drug therapy , Granuloma/pathology , Male , Mitogen-Activated Protein Kinase 3/metabolism , Palmitic Acids/therapeutic use , Phosphorylation/drug effects , Rats , Rats, Wistar , Transcription, Genetic/drug effects
6.
PLoS One ; 7(10): e47464, 2012.
Article in English | MEDLINE | ID: mdl-23077623

ABSTRACT

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.


Subject(s)
Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Indazoles/administration & dosage , Monocyte Chemoattractant Proteins/metabolism , Propionates/administration & dosage , Actins/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cell Movement/physiology , Coronary Vessels/cytology , Coronary Vessels/drug effects , Gene Expression/drug effects , Humans , Microfilament Proteins/metabolism , Monocyte Chemoattractant Proteins/antagonists & inhibitors , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myosin Heavy Chains/metabolism , Neointima/pathology , Rats , Calponins
7.
PLoS One ; 6(12): e28159, 2011.
Article in English | MEDLINE | ID: mdl-22163000

ABSTRACT

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.


Subject(s)
Cannabidiol/pharmacology , Inflammation/metabolism , Intestines/drug effects , Animals , Biopsy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/immunology , Colon/metabolism , Humans , Immune System , Inflammation/drug therapy , Interferon-gamma/metabolism , Lipopolysaccharides/metabolism , Macrophages/cytology , Male , Mast Cells/cytology , Mice , Nervous System/metabolism , Nitrites/metabolism , Sepsis
8.
PLoS One ; 6(12): e28668, 2011.
Article in English | MEDLINE | ID: mdl-22163051

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Cannabidiol/pharmacology , Hippocampus/metabolism , Neurogenesis , PPAR gamma/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Binding Sites , Brain/metabolism , Inflammation/metabolism , Male , Neurons/metabolism , Neurons/pathology , Nitric Oxide/metabolism , Protein Binding , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Time Factors
9.
J Cell Mol Med ; 15(12): 2664-74, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21255263

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Astrocytes/drug effects , Gliosis/drug therapy , Inflammation/drug therapy , Palmitic Acids/pharmacology , Peptide Fragments/adverse effects , Amides , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Blotting, Western , Electrophoretic Mobility Shift Assay , Endocannabinoids , Ethanolamines , Fluorescent Antibody Technique , Gliosis/etiology , Humans , Inflammation/etiology , PPAR alpha/antagonists & inhibitors , PPAR alpha/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
10.
Mol Pain ; 7: 3, 2011 Jan 10.
Article in English | MEDLINE | ID: mdl-21219627

ABSTRACT

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.


Subject(s)
Granuloma/complications , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Mast Cells/pathology , Palmitic Acids/therapeutic use , Amides , Animals , Carrageenan , Endocannabinoids , Ethanolamines , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Granuloma/pathology , Hyperalgesia/pathology , Male , Mast Cells/drug effects , Nerve Fibers/drug effects , Nerve Fibers/pathology , Nerve Growth Factors/metabolism , Neurogenesis/drug effects , Palmitic Acids/administration & dosage , Palmitic Acids/pharmacology , Rats , Rats, Wistar , Time Factors
11.
CNS Neurol Disord Drug Targets ; 9(6): 807-12, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20874699

ABSTRACT

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.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neurodegenerative Diseases/drug therapy , Animals , Brain/blood supply , Brain/drug effects , Humans , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Neurodegenerative Diseases/physiopathology
12.
Endocrinology ; 151(3): 921-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20133454

ABSTRACT

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.


Subject(s)
Arachidonic Acids/metabolism , Carcinoma/metabolism , Endometrial Neoplasms/metabolism , Glycerides/metabolism , Receptor, Cannabinoid, CB2/metabolism , Adult , Biopsy , Carcinoma/pathology , Cell Line, Tumor , Down-Regulation , Endocannabinoids , Endometrial Neoplasms/pathology , Endometrium/pathology , Female , Humans , Middle Aged , Monoacylglycerol Lipases/metabolism , Up-Regulation
13.
ScientificWorldJournal ; 9: 229-35, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19347234

ABSTRACT

Activated microglia and astrocytes produce a large number of inflammatory and neurotoxic substances in various brain pathologies, above all during neurodegenerative disorders. In the search for new neuroprotective compounds, interest has turned to marijuana derivatives, since in several in vitro, in vivo, and clinical studies, they have shown a great ability to control neuroinflammation. Despite the emerging evidence regarding pharmacological activities of cannabinoids, their effective introduction into clinical therapy still remains controversial and strongly limited by their unavoidable psychotropicity. Since the psychotropic effect of cannabinoids is generally linked to the activation of the CB1 receptor on neurons, the aim of our review is to clarify the function of the two cannabinoid receptors on glial cells and the differential role played by them, highlighting the emerging evidence of a CB2-mediated control of neuroinflammation that could liberate cannabinoids from the slavery of their central side effects. Despite the emerging evidence regarding pharmacological activities of cannabinoids, however their effective introduction in the clinical therapy remains still controversial and strongly limited by their unavoidable psychotropicity. Since the psychotropic effect of cannabinoids is generally linked to the activation of CB1 receptor on neurons, aim of our review is to clarify the functioning of the two cannabinoid receptors on glial cells and the differential role played by them, highlighting the emerging evidence of a CB2-mediated control of neuro-inflammation that could liberate cannabinoids from the slavery of the central side effects.


Subject(s)
Cannabinoids/pharmacology , Gliosis/metabolism , Microglia/physiology , Neuroprotective Agents/pharmacology , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB2/drug effects , Animals , Cannabinoids/therapeutic use , Gene Expression , Gliosis/drug therapy , Humans , Microglia/drug effects , Receptor, Cannabinoid, CB1/physiology , Receptor, Cannabinoid, CB2/physiology
14.
CNS Neurosci Ther ; 15(1): 65-75, 2009.
Article in English | MEDLINE | ID: mdl-19228180

ABSTRACT

Neurodegenerative diseases represent, nowadays, one of the main causes of death in the industrialized country. They are characterized by a loss of neurons in particular regions of the nervous system. It is believed that this nerve cell loss underlies the subsequent decline in cognitive and motor function that patients experience in these diseases. A range of mutant genes and environmental toxins have been implicated in the cause of neurodegenerative disorders but the mechanism remains largely unknown. At present, inflammation, a common denominator among the diverse list of neurodegenerative diseases, has been implicated as a critical mechanism that is responsible for the progressive nature of neurodegeneration. Since, at present, there are few therapies for the wide range of neurodegenerative diseases, scientists are still in search of new therapeutic approaches to the problem. An early contribution of neuroprotective and antiinflammatory strategies for these disorders seems particularly desirable because isolated treatments cannot be effective. In this contest, marijuana derivatives have attracted special interest, although these compounds have always raised several practical and ethical problems for their potential abuse. Nevertheless, among Cannabis compounds, cannabidiol (CBD), which lacks any unwanted psychotropic effect, may represent a very promising agent with the highest prospect for therapeutic use.


Subject(s)
Cannabidiol/therapeutic use , Neurodegenerative Diseases/drug therapy , Alzheimer Disease/drug therapy , Animals , Cannabidiol/metabolism , Cannabidiol/pharmacology , Cytoprotection , Disease Models, Animal , Humans , Huntington Disease/drug therapy , Multiple Sclerosis/drug therapy , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Prion Diseases/drug therapy
15.
J Cell Mol Med ; 13(6): 1086-95, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18429935

ABSTRACT

Palmitoylethanolamide (PEA) and some of its analogues have shown great efficacy in the treatment of pain and inflammation. Adelmidrol - the International Nonproprietary Name (INN) of the di-amide derivative of azelaic acid - is one of these analogues. The anti-inflammatory and analgesic effects of PEA and adelmidrol are hypothesized to be mediated, at least in part, by mast cell down-modulation. Mast cell mediators released at early stage of the inflammatory process drive the inflammatory reaction to chronicity as it happens in X-carrageenin-induced granulomatous tissue formation. In the present study, the choice of testing adelmidrol depends upon the physicochemical properties of the compound, i.e. the amphipatic feature, that make it more easily soluble than PEA. In this study, we investigated the effect of adelmidrol on granuloma formation induced by lambda-carrageenin-soaked sponge implant in rats. Our results show that the local administration of the compound under study significantly decreases weight and neo-angiogenesis in granulomatous tissue. The anti-inflammatory effect was due to the modulation of mast cells degranulation, as shown by histological analysis and by the inhibition of the release of several pro-inflammatory and pro-angiogenic enzymes (e.g. iNOS, chymase and metalloproteinase MMP-9), and mediators (e.g. nitric oxide and TNF-alpha). The results indicate that adelmidrol, given locally, may represent a potential therapeutic tool in controlling chronic inflammation.


Subject(s)
Dicarboxylic Acids/pharmacology , Granuloma/prevention & control , Inflammation/prevention & control , Palmitic Acids/pharmacology , Amides , Animals , Blotting, Western , Carrageenan , Cell Count , Chronic Disease , Dicarboxylic Acids/chemistry , Dose-Response Relationship, Drug , Endocannabinoids , Ethanolamines , Granuloma/chemically induced , Granuloma/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Leukocytes/drug effects , Leukocytes/pathology , Male , Mast Cells/drug effects , Mast Cells/pathology , Neovascularization, Pathologic/prevention & control , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Palmitic Acids/chemistry , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
16.
J Cell Mol Med ; 12(3): 914-27, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18494933

ABSTRACT

Previous studies suggest that levels of the astrocyte-derived S100B protein, such as those occurring in brain extra-cellular spaces consequent to persistent astroglial activation, may have a pathogenetic role in Alzheimer's disease (AD). Although S100B was reported to promote beta amyloid precursor protein overexpression, no clear mechanistic relationship between S100B and formation of neurofibrillary tangles (NFTs) is established. This in vitro study has been aimed at investigating whether S100B is able to disrupt Wnt pathway and lead to tau protein hyperphosphorylation. Utilizing Western blot, electrophoretic mobility shift assay, supershift and reverse transcriptase-polymerase chain reaction techniques, it has been demonstrated that micromolar S100B concentrations stimulate c-Jun N-terminal kinase (JNK) phosphorylation through the receptor for advanced glycation ending products, and subsequently activate nuclear AP-1/cJun transcription, in cultured human neural stem cells. In addition, as revealed by Western blot, small interfering RNA and immunofluorescence analysis, S100B-induced JNK activation increased expression of Dickopff-1 that, in turn, promoted glycogen synthase kinase 3beta phosphorylation and beta-catenin degradation, causing canonical Wnt pathway disruption and tau protein hyperphosphorylation. These findings propose a previously unrecognized link between S100B and tau hyperphosphorylation, suggesting S100B can contribute to NFT formation in AD and in all other conditions in which neuroinflammation may have a crucial role.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Nerve Growth Factors/pharmacology , Neurons/metabolism , S100 Proteins/pharmacology , Stem Cells/metabolism , Up-Regulation , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cells, Cultured , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Neurofibrillary Tangles/pathology , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , S100 Calcium Binding Protein beta Subunit , Time Factors , Wnt Proteins/metabolism
17.
J Cell Mol Med ; 12(2): 661-70, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18419603

ABSTRACT

Endometritis is defined as an inflammation of the endometrial mucosa of the uterus. In endometritis large amounts of toxic mediators, including nitric oxide (NO) are released by inflammatory cells. As a consequence of nitric oxide-dependent injury, the cells respond by triggering protective mechanisms, by changing the endocannabinoid system (ECS) which comprises both CB(1) and CB(2) cannabinoid receptors and their endogenous ligands. The aim of our study was to seek out evidence for the presence of cannabinoid receptors in inflammatory endometrial tissue as well as for their potential role in endometrial inflammation. Our results showed a selective up-regulation of both transcription and expression of CB(2) receptors in biopsies from women affected by endometrial inflammation compared to healthy women. The experiments with the nitric oxide-donor S-Nitroso-L-Glutathione (GSNO) suggest that such a selective up-regulation may be related to the nitric oxide release occurring during endometrial inflammation. In addition, we demonstrated an increase in chymase expression, a marker of mast cells, in biopsies of women affected by endometritis. Therefore our results support the hypothesis that the up-regulation of CB(2) occurs mainly on mast cells and that it might tend to sensitize these cells to the anti-inflammatory effect exerted by endogenous cannabinoids by binding their receptor and thus preventing the mast cell degranulation and the release of pro-inflammatory mediators. In conclusion, we believe that the selective CB(2) up-regulation might play a role as a novel prognostic factor in endometrial inflammation.


Subject(s)
Endometritis/pathology , Endometritis/physiopathology , Receptor, Cannabinoid, CB2 , Up-Regulation , Biopsy , Calcimycin/pharmacology , Case-Control Studies , Cell Degranulation/drug effects , Chymases/metabolism , Endometritis/surgery , Female , Humans , Indoles/pharmacology , Ionophores/pharmacology , Mast Cells/physiology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type II/metabolism , Nitrites/analysis , Organ Culture Techniques , Peroxidase/metabolism , RNA, Messenger/analysis , S-Nitrosoglutathione/pharmacology , Transcription, Genetic , beta-N-Acetylhexosaminidases/analysis
18.
J Pineal Res ; 44(1): 45-51, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18078447

ABSTRACT

A growing number of reports demonstrate that a pro-inflammatory and oxidative condition is related to the pathogenesis and the progression of endotoxin-induced septic shock and that antioxidants may have therapeutic potential in lipopolysaccharide (LPS)-induced sepsis. Melatonin has been shown to possess potent antioxidant properties in several models of inflammation in mice and rats. In the present study we focused on the possible protective mechanism of melatonin in preventing gastrointestinal (GI) disturbances induced by LPS in mice. In fact, mice treated with LPS showed a reduced gastric emptying of solid beads. Also the geometric center, representing the relative distribution of the solid beads throughout the entire GI tract, was significantly reduced in LPS-treated mice confirming that sepsis leads to a disturbed GI motility in mice. Melatonin completely reversed the LPS-induced motility disturbance. This beneficial effect of melatonin is associated with a reduction in lipid peroxidation, MAPK activation, NF-kappaB activation, iNOS transcription and expression and nitrite production in intestinal tissue from septic mice. These results demonstrate that melatonin prevents the LPS-induced GI disturbances in mice switching off the pro-oxidant pathways induced by the endotoxin. Therefore, it is reasonable to propose melatonin as a molecule with therapeutic potential for the treatment of systemic inflammation by interfering at the earliest steps of activation of the oxidative and pro-inflammatory cascade.


Subject(s)
Antioxidants/pharmacology , Gastroenteritis/drug therapy , Gastrointestinal Motility/drug effects , Melatonin/pharmacology , Oxidative Stress/drug effects , Animals , Antioxidants/therapeutic use , Gastroenteritis/chemically induced , Lipopolysaccharides/toxicity , Male , Melatonin/therapeutic use , Mice
19.
Hum Mol Genet ; 17(3): 440-57, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17984171

ABSTRACT

Down syndrome (DS) is caused by trisomy of chromosome 21 and is characterized by mental retardation, seizures and premature Alzheimer's disease. To examine neuropathological mechanisms giving rise to this disorder, we generated multiple human DS neural progenitor cell (NPC) lines from the 19-21 week frontal cortex and characterized their genomic and functional properties. Microarray profiling of DS progenitors indicated that increased levels of gene expression were not limited to chromosome 21, suggesting that increased expression of genes on chromosome 21 altered transcriptional regulation of a subset of genes throughout the entire genome. Moreover, many transcriptionally dysregulated genes were involved in cell death and oxidative stress. Network analyses suggested that upregulated expression of chromosome 21 genes such as S100B and amyloid precursor protein activated the stress response kinase pathways, and furthermore, could be linked to upregulation of the water channel aquaporin 4 (AQP4). We further demonstrate in DS NPCs that S100B is constitutively overexpressed, that overexpression leads to increased reactive oxygen species (ROS) formation and activation of stress response kinases, and that activation of this pathway results in compensatory AQP4 expression. In addition, AQP4 expression could be induced by direct exposure to ROS, and siRNA inhibition of AQP4 resulted in elevated levels of ROS following S100B exposure. Finally, elevated levels of S100B-induced ROS and loss of AQP4 expression led to increased programmed cell death. These findings suggest that dysregulation of chromosome 21 genes in DS neural progenitors leads to increased ROS and thereby alters transcriptional regulation of cytoprotective, non-chromosome 21 genes in response to ongoing cellular insults.


Subject(s)
Aquaporin 4/genetics , Aquaporin 4/metabolism , Down Syndrome/genetics , Down Syndrome/metabolism , Embryonic Stem Cells/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neurons/metabolism , S100 Proteins/genetics , S100 Proteins/metabolism , Apoptosis , Base Sequence , Case-Control Studies , Cell Line , Chromosomes, Human, Pair 21/genetics , DNA Primers/genetics , Down Syndrome/embryology , Down Syndrome/pathology , Embryonic Stem Cells/pathology , Gene Expression Profiling , Humans , Neurons/pathology , Oligonucleotide Array Sequence Analysis , Oxidative Stress , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , S100 Calcium Binding Protein beta Subunit , Transfection
20.
Anticancer Res ; 27(5A): 3083-9, 2007.
Article in English | MEDLINE | ID: mdl-17970048

ABSTRACT

BACKGROUND: Mammary tumors are a very common neoplasm in the dog and show histological features and biological behaviour similar to human mammary carcinomas. Recently, a pathway named Wnt-1, involving beta-catenin and APC protein, has emerged as an important player in many human tumor types, including mammary neoplasms. MATERIALS AND METHODS: Thirty-five samples of canine mammary tumors (10 benign and 25 malignant) were studied in order to evaluate the co-expression of beta-catenin, APC protein and E cadherin with confocal laser microscopical observation, by western blot analysis and by correlating data obtained with the histological grade of tumours. RESULTS: A progressive decrease of E-cadherin together with disruption of beta-catenin expression was observed in less differentiated malignant tumors. In addition, a loss of beta-catenin membranous distribution and a cytoplasmic accumulation was often coexpressed with disrupted expression of APC protein. Western blot analysis showed a progressive increase of beta-catenin in malignant tumors, which could be the expression of disrupted /-catenin catabolism leading to cytoplasmic accumulation. In some less differentiated malignant tumors, a marked beta-catenin decrease was also observed. This feature could be linked to mutations in beta-catenin gene coding for a truncated and lighter protein. CONCLUSION: These results may indicate the multifunctional role played by beta-catenin in canine mammary oncogenesis.


Subject(s)
Adenocarcinoma/metabolism , Adenomatous Polyposis Coli Protein/biosynthesis , Cadherins/biosynthesis , Dog Diseases/metabolism , Mammary Neoplasms, Animal/metabolism , beta Catenin/biosynthesis , Adenocarcinoma/pathology , Animals , Blotting, Western , Dog Diseases/pathology , Dogs , Fluorescent Antibody Technique , Mammary Neoplasms, Animal/pathology , Microscopy, Confocal
SELECTION OF CITATIONS
SEARCH DETAIL
...