Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(13)2023 Jun 27.
Article in English | MEDLINE | ID: mdl-37445884

ABSTRACT

In a previous study, we showed that various low-molecular-weight compounds in follicular fluid (FF) samples of control fertile females (CFF) have different concentrations compared to those found in FF of infertile females (IF), before and after their categorization into different subgroups, according to their clinical diagnosis of infertility. Using the same FF samples of this previous study, we here analyzed the FF concentrations of free and bound bilirubin and compared the results obtained in CFF, IF and the different subgroups of IF (endometriosis, EM, polycystic ovary syndrome, PCOS, age-related reduced ovarian reserve, AR-ROR, reduced ovarian reserve, ROR, genetic infertility, GI and unexplained infertility, UI). The results clearly indicated that CFF had lower values of free, bound and total bilirubin compared to the respective values measured in pooled IF. These differences were observed even when IF were categorized into EM, PCOS, AR-ROR, ROR, GI and UI, with EM and PCOS showing the highest values of free, bound and total bilirubin among the six subgroups. Using previous results of ascorbic acid, GSH and nitrite + nitrate measured in the same FF samples of the same FF donors, we found that total bilirubin in FF increased as a function of decreased values of ascorbic acid and GSH, and increased concentrations of nitrite + nitrate. The values of total bilirubin negatively correlated with the clinical parameters of fertilization procedures (number of retrieved oocytes, mature oocytes, fertilized oocytes, blastocysts, high-quality blastocysts) and with clinical pregnancies and birth rates. Bilirubin concentrations in FF were not linked to those found in serum samples of FF donors, thereby strongly suggesting that its over production was due to higher activity of heme oxygenase-1 (HO-1), the key enzyme responsible for bilirubin formation, in granulosa cells, or cumulus cells or oocytes of IF and ultimately leading to bilirubin accumulation in FF. Since increased activity of HO-1 is one of the main enzymatic intracellular mechanisms of defense towards external insults (oxidative/nitrosative stress, inflammation), and since we found correlations among bilirubin and oxidative/nitrosative stress in these FF samples, it may reasonably be supposed that bilirubin increase in FF of IF is the result of protracted exposures to the aforementioned insults evidently playing relevant roles in female infertility.


Subject(s)
Infertility, Female , Polycystic Ovary Syndrome , Pregnancy , Humans , Female , Infertility, Female/metabolism , Follicular Fluid/metabolism , Antioxidants/metabolism , Nitric Oxide/metabolism , Polycystic Ovary Syndrome/metabolism , Nitrates/metabolism , Nitrites/metabolism , Fertilization in Vitro , Oocytes/metabolism , Outcome Assessment, Health Care , Bilirubin/metabolism , Ascorbic Acid/metabolism
2.
Int J Mol Sci ; 24(12)2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37373229

ABSTRACT

Endometrial hyperplasia is a threatening pathology driven by unopposed estrogen stimulus. Moreover, insulin may act on the endometrium, prompting further growth. We aimed at assessing whether D-chiro-Inositol, an insulin sensitizer with estrogen-lowering properties, might improve the condition of patients with simple endometrial hyperplasia without atypia. We enrolled women with simple endometrial hyperplasia without atypia and related symptoms, including abnormal uterine bleeding. We treated the patients with one tablet per day, containing 600 mg of D-chiro-inositol for six months. Patients underwent ultrasound to assess the thickness of the endometrium at baseline, after three months, and at the end of this study. Endometrial thickness went from 10.82 ± 1.15 mm to 8.00 ± 0.81 mm after three months (p < 0.001) and to 6.9 ± 1.06 mm after six months (p < 0.001 versus baseline; p < 0.001 versus three months). D-chiro-inositol treatment also improved heavy menstrual bleeding and the length of menstruation. Despite the fact that our data should be validated in larger studies with appropriate control groups, our promising results support the hypothesis that D-chiro-inositol may represent a useful treatment in the case of endometrial hyperplasia without atypia.


Subject(s)
Endometrial Hyperplasia , Insulins , Female , Humans , Endometrial Hyperplasia/drug therapy , Endometrial Hyperplasia/pathology , Pilot Projects , Inositol/therapeutic use , Endometrium/pathology , Estrogens
3.
Int J Mol Sci ; 24(1)2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36614333

ABSTRACT

Notwithstanding the great improvement of ART, the overall rate of successful pregnancies from implanted human embryos is definitely low. The current routine embryo quality assessment is performed only through morphological criteria, which has poor predictive capacity since only a minor percentage of those in the highest class give rise to successful pregnancy. Previous studies highlighted the potentiality of the analysis of metabolites in human embryo culture media, useful for the selection of embryos for implantation. In the present study, we analyzed in blind 66 human embryo culture media at 5 days after in vitro fertilization with the aim of quantifying compounds released by cell metabolism that were not present as normal constituents of the human embryo growth media, including purines, pyrimidines, nitrite, and nitrate. Only some purines were detectable (hypoxanthine and uric acid) in the majority of samples, while nitrite and nitrate were always detectable. When matching biochemical results with morphological evaluation, it was found that low grade embryos (n = 12) had significantly higher levels of all the compounds of interest. Moreover, when matching biochemical results according to successful (n = 17) or unsuccessful (n = 25) pregnancy, it was found that human embryos from the latter group released higher concentrations of hypoxanthine, uric acid, nitrite, and nitrate in the culture media. Additionally, those embryos that developed into successful pregnancies were all associated with the birth of healthy newborns. These results, although carried out on a relatively low number of samples, indicate that the analysis of the aforementioned compounds in the culture media of human embryos is a potentially useful tool for the selection of embryos for implantation, possibly leading to an increase in the overall rate of ART.


Subject(s)
Embryo Transfer , Nitric Oxide , Infant, Newborn , Pregnancy , Female , Humans , Culture Media/metabolism , Nitrates , Nitrites , Uric Acid , Embryo Implantation , Fertilization in Vitro , Energy Metabolism , Hypoxanthines , Embryo Culture Techniques , Pregnancy Rate
4.
J Clin Med ; 12(1)2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36615188

ABSTRACT

Background: Recent studies reported possible concerns following long-lasting treatments with high doses of D-chiro-inositol in women. However, to date, no clinical trial has investigated or validated these concerns. We addressed this issue both retrospectively and with a prospective pilot study. Methods: For the retrospective analysis, we searched our databases for insulin-resistant women who took 1200 mg/day D-chiro-inositol for 6 months. In our prospective study, we enrolled 10 healthy women to supplement with the same therapeutic scheme. We performed statistical analyses through the Wilcoxon Signed-Rank Test. A p-value < 0.05 was considered significant. Results: Twenty women underwent 6 months of 1200 mg/day D-chiro-inositol. The treatment significantly decreased BMI, glycemia, insulinemia, HOMA-IR, serum levels of LH, total testosterone, and DHEAS. Serum estradiol rose and menstrual abnormalities occurred following the treatment. In our prospective study, we observed increases in serum levels of total testosterone and asprosin in healthy women. Conclusions: This is the first clinical evidence demonstrating that long-term treatments with high dosages of D-chiro-inositol can predispose women to hormonal and menstrual abnormalities. Moreover, the accumulation of D-chiro-inositol following such treatment regimen may lead to detrimental effects in non-reproductive tissues, as demonstrated by the increase in asprosin levels.

5.
Sci Rep ; 7(1): 7735, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28798420

ABSTRACT

Despite the effectiveness of combined anti-retroviral therapy, human immunodeficiency virus (HIV) infected-patients frequently report diarrhea and neuropsychological deficits. It is claimed that the viral HIV-1 Trans activating factor (HIV-1 Tat) protein is responsible for both diarrhea and neurotoxic effects, but the underlying mechanisms are not known. We hypothesize that colonic application of HIV-1 Tat activates glial cells of the enteric nervous system (EGCs), leading to a neuroinflammatory response able to propagate to the central nervous system. We demonstrated that HIV-1 Tat-induced diarrhea was associated with a significant activation of glial cells within the colonic wall, the spinal cord and the frontal cortex, and caused a consistent impairment of the cognitive performances. The inhibition of glial cells activity by lidocaine, completely abolished the above-described effects. These observations point out the role of glial cells as putative effectors in HIV-1 Tat-associated gastrointestinal and neurological manifestations and key regulators of gut-brain signaling.


Subject(s)
Central Nervous System/metabolism , Central Nervous System/physiopathology , Diarrhea/etiology , Enteric Nervous System/metabolism , Enteric Nervous System/physiopathology , Inflammation/etiology , Neuroglia/metabolism , tat Gene Products, Human Immunodeficiency Virus/adverse effects , Animals , Biomarkers , Central Nervous System/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Cognition Disorders/etiology , Cognition Disorders/metabolism , Cognition Disorders/psychology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Gliosis , HIV Infections/complications , HIV Infections/virology , Inflammation/metabolism , Male , Rats , S100 Calcium Binding Protein beta Subunit/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/physiopathology
6.
Front Cell Neurosci ; 11: 42, 2017.
Article in English | MEDLINE | ID: mdl-28280459

ABSTRACT

S100B is expressed in various types of glial cells and is involved in regulating many aspects of their function. However, little is known about its role during nervous system development. In this study, we investigated the effect of inhibiting the onset of S100B synthesis in the development of the enteric nervous system, a network of neurons and glia located in the wall of the gut that is vital for control of gastrointestinal function. Intact gut explants were taken from embryonic day (E)13.5 mice, the day before the first immunohistochemical detection of S100B, and cultured in the presence of arundic acid, an inhibitor of S100B synthesis, for 48 h. The effects on Sox10-immunoreactive enteric neural crest progenitors and Hu-immunoreactive enteric neurons were then analyzed. Culture in arundic acid reduced the proportion of Sox10+ cells and decreased cell proliferation. There was no change in the density of Hu+ enteric neurons, however, a small population of cells exhibited atypical co-expression of both Sox10 and Hu, which was not observed in control cultures. Addition of exogenous S100B to the cultures did not change Sox10+ cell numbers. Overall, our data suggest that cell-intrinsic intracellular S100B is important for maintaining Sox10 and proliferation of the developing enteric glial lineage.

7.
Front Pharmacol ; 7: 120, 2016.
Article in English | MEDLINE | ID: mdl-27242527

ABSTRACT

BACKGROUND: Clostridium difficile infections (CDIs) caused by Clostridium difficile toxin A (TcdA) lead to severe ulceration, inflammation and bleeding of the colon, and are difficult to treat. AIM: The study aimed to evaluate the effect of rifaximin on TcdA-induced apoptosis in intestinal epithelial cells and investigate the role of PXR in its mechanism of action. METHODS: Caco-2 cells were incubated with TcdA and treated with rifaximin (0.1-10 µM) with or without ketoconazole (10 µM). The transepithelial electrical resistance (TEER) and viability of the treated cells was determined. Also, the expression of zona occludens-1 (ZO-1), toll-like receptor 4 (TLR4), Bcl-2-associated X protein (Bax), transforming growth factor-ß-activated kinase-1 (TAK1), myeloid differentiation factor 88 (MyD88), and nuclear factor-kappaB (NF-κB) was determined. RESULTS: Rifaximin treatment (0.1, 1.0, and 10 µM) caused a significant and concentration-dependent increase in the TEER of Caco-2 cells (360, 480, and 680% vs. TcdA treatment) 24 h after the treatment and improved their viability (61, 79, and 105%). Treatment also concentration-dependently decreased the expression of Bax protein (-29, -65, and -77%) and increased the expression of ZO-1 (25, 54, and 87%) and occludin (71, 114, and 262%) versus TcdA treatment. The expression of TLR4 (-33, -50, and -75%), MyD88 (-29, -60, and -81%) and TAK1 (-37, -63, and -79%) were also reduced with rifaximin versus TcdA treatment. Ketoconazole treatment inhibited these effects. CONCLUSION: Rifaximin improved TcdA-induced toxicity in Caco-2 cells by the PXR-dependent TLR4/MyD88/NF-κB pathway mechanism, and may be useful in the treatment of CDIs.

8.
Int J Oncol ; 49(2): 639-45, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27279570

ABSTRACT

Activation of intestinal human pregnane X receptor (PXR) has recently been proposed as a promising strategy for the chemoprevention of inflammation-induced colon cancer. The present study was aimed at evaluating the effect of rifaximin, a non-absorbable antibiotic, in inhibiting angiogenesis in a model of human colorectal epithelium and investigating the role of PXR in its mechanism of action. Caco-2 cells were treated with rifaximin (0.1, 1.0 and 10.0 µM) in the presence or absence of ketoconazole (10 µM) and assessed for cell proliferation, migration and expression of proliferating cell nuclear antigen (PCNA). The release of vascular endothelial growth factor (VEGF) and nitric oxide (NO), expression of Akt, mechanistic target of rapamycin (mTOR), p38 mitogen activated protein kinases (MAPK), nuclear factor κB (NF-κB) and metalloproteinase-2 and -9 (MMP-2 and -9) were also evaluated. Treatment with rifaximin 0.1, 1.0 and 10.0 µM caused significant and concentration-dependent reduction of cell proliferation, cell migration and PCNA expression in the Caco-2 cells vs. untreated cells. Treatment downregulated VEGF secretion, NO release, VEGFR-2 expression, MMP-2 and MMP-9 expression vs. untreated cells. Rifaximin treatment also resulted in a concentration-dependent decrease in the phosphorylation of Akt, mTOR, p38MAPK and inhibition of hypoxia-inducible factor 1-α (HIF-1α), p70S6K and NF-κB. Ketoconazole (PXR antagonist) treatment inhibited these effects. These findings demonstrated that rifaximin causes PXR-mediated inhibition of angiogenic factors in Caco-2 cell line and may be a promising anticancer tool.


Subject(s)
Colonic Neoplasms/drug therapy , Inflammation/drug therapy , Neovascularization, Pathologic/drug therapy , Receptors, Steroid/biosynthesis , Rifamycins/administration & dosage , Anti-Bacterial Agents/administration & dosage , Caco-2 Cells , Cell Movement/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/etiology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Ketoconazole/administration & dosage , Neoplasm Proteins/biosynthesis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Pregnane X Receptor , Proliferating Cell Nuclear Antigen/biosynthesis , Receptors, Steroid/antagonists & inhibitors , Rifaximin , Signal Transduction/drug effects
9.
PLoS One ; 11(5): e0156198, 2016.
Article in English | MEDLINE | ID: mdl-27219328

ABSTRACT

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.


Subject(s)
Colitis, Ulcerative/drug therapy , Colitis/drug therapy , Dextran Sulfate/adverse effects , Ethanolamines/administration & dosage , PPAR alpha/metabolism , Palmitic Acids/administration & dosage , Vascular Endothelial Growth Factor A/metabolism , Adult , Amides , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/metabolism , Colitis, Ulcerative/metabolism , Disease Models, Animal , Ethanolamines/pharmacology , Female , Humans , Male , Mice , Middle Aged , Palmitic Acids/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
10.
Phytother Res ; 30(6): 963-70, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26929026

ABSTRACT

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.


Subject(s)
Colonic Neoplasms/drug therapy , Ethanolamines/chemistry , PPAR alpha/metabolism , Palmitic Acids/chemistry , Vascular Endothelial Growth Factor A/metabolism , Amides , Animals , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation , Down-Regulation/drug effects , Ethanolamines/therapeutic use , Humans , Neovascularization, Pathologic , Palmitic Acids/therapeutic use , Signal Transduction
11.
Sci Rep ; 6: 22605, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26940982

ABSTRACT

Alzheimer's disease (AD) is characterized by chronic deposition of ß-amyloid (Aß) in the brain, progressive neurodegeneration and consequent cognitive and behavioral deficits that typify the disease. Astrocytes are pivotal in this process because they are activated in the attempt to digest Aß which starts a neuroinflammatory response that further contributes to neurodegeneration. The intestine is a good source of astrocytes-like cells-referred to as enteric glial cells (EGCs). Here we show that the autologous transplantation of EGCs into the brain of Aß-injected rats arrested the development of the disease after their engraftment. Transplanted EGCs showed anti-amyloidogenic activity, embanked Aß-induced neuroinflammation and neurodegeneration, and released neutrophic factors. The overall result was the amelioration of the pathological hallmarks and the cognitive and behavioral deficits typical of Aß-associated disease. Our data indicate that autologous EGCs transplantation may provide an efficient alternative for applications in cell-replacement therapies to treat neurodegeneration in AD.


Subject(s)
Cognition Disorders/therapy , Intestines/pathology , Neurodegenerative Diseases/therapy , Neuroglia/transplantation , Transplantation, Autologous , Amyloid beta-Peptides/administration & dosage , Animals , Cells, Cultured , Cognition , Cognition Disorders/chemically induced , Disease Models, Animal , Humans , Male , Neurodegenerative Diseases/chemically induced , Neuroglia/pathology , Rats , Rats, Sprague-Dawley , Recovery of Function
12.
Hum Mol Genet ; 25(9): 1714-27, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26911678

ABSTRACT

Down syndrome (DS) is caused by a triplication of chromosome 21 (HSA21). Increased oxidative stress, decreased neurogenesis and synaptic dysfunction from HSA21 gene overexpression are thought to cause mental retardation, dementia and seizure in this disorder. Recent epigenetic studies have raised the possibility that DNA methylation has significant effects on DS neurodevelopment. Here, we performed methylome profiling in normal and DS fetal cortices and observed a significant hypermethylation in ∼4% of probes in the DS samples compared with age-matched normals. The probes with differential methylation were distributed across all chromosomes, with no enrichment on HSA21. Functional annotation and pathway analyses showed that genes in the ubiquitination pathway were significantly altered, including: BRCA1, TSPYL5 and PEX10 HSA21 located DNMT3L was overexpressed in DS neuroprogenitors, and this overexpression increased the promoter methylation of TSPYL5 potentially through DNMT3B, and decreased its mRNA expression. DNMT3L overexpression also increased mRNA levels for TP53 and APP, effectors of TSPYL5 Furthermore, DNMT3L overexpression increased APP and PSD95 expression in differentiating neurons, whereas DNMT3LshRNA could partially rescue the APP and PSD95 up-regulation in DS cells. These results provide some of the first mechanistic insights into causes for epigenetic changes in DS, leading to modification of genes relevant for the DS neural endophenotype.


Subject(s)
Cerebral Cortex/metabolism , Chromosomes, Human, Pair 21/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Down Syndrome/genetics , Epigenesis, Genetic/genetics , Fetus/metabolism , Biomarkers/metabolism , Blood Proteins/genetics , Case-Control Studies , Cells, Cultured , Cerebral Cortex/pathology , DNA Methylation , Disks Large Homolog 4 Protein , Fetus/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Nuclear Proteins/genetics , Oxidative Stress , Poly(A)-Binding Proteins/genetics , Promoter Regions, Genetic , Ubiquitination
13.
Biomed Res Int ; 2015: 508342, 2015.
Article in English | MEDLINE | ID: mdl-26295040

ABSTRACT

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.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Gliosis/complications , Gliosis/drug therapy , Neurons/pathology , Pentamidine/therapeutic use , S100 Calcium Binding Protein beta Subunit/antagonists & inhibitors , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Densitometry , Dinoprostone/metabolism , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Hippocampus/drug effects , Hippocampus/pathology , Humans , Immunohistochemistry , Inflammation/complications , Inflammation/pathology , Injections , Interleukin-1beta/metabolism , Malondialdehyde/metabolism , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Nitrites/metabolism , Pentamidine/pharmacology , S100 Calcium Binding Protein beta Subunit/metabolism , Tumor Suppressor Protein p53/metabolism
14.
Oncol Lett ; 9(6): 2864-2870, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26137161

ABSTRACT

S100 calcium-binding protein B (S100B) is highly expressed in glioma cells and promotes cancer cell survival via inhibition of the p53 protein. In melanoma cells, this S100B-p53 interaction is known to be inhibited by pentamidine isethionate, an antiprotozoal agent. Thus, the aim of the present study was to evaluate the effect of pentamidine on rat C6 glioma cell proliferation, migration and apoptosis in vitro. The change in C6 cell proliferation following treatment with pentamidine was determined by performing a 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyltetrazolium bromide-formazan assay. Significant dose-dependent decreases in proliferation were observed at pentamidine concentrations of 0.05 µM (58.5±5%; P<0.05), 0.5 µM (40.6±7%; P<0.01) and 5 µM (13±4%; P<0.001) compared with the control (100% viability). Furthermore, treatment with 0.05, 0.5 and 5 µM pentamidine was associated with a significant increase in apoptosis versus the untreated cells, as determined by DNA fragmentation assays, immunofluorescence analysis of C6 chromatin using Hoechst staining, and immunoblot analysis of B-cell lymphoma-2 (Bcl-2)-associated X protein (100%, P<0.05; 453%, P<0.01; and 1000%, P<0.001, respectively) and Bcl-2 (-60%, P<0.001; -80.13%, P<0.001; -95%, P<0.001, respectively). In addition, the administration of 0.05, 0.5 and 5 µM pentamidine significantly upregulated the protein expression levels of p53 (681±87.5%, P<0.05; 1244±94.3%, P<0.01; and 2244±111%, P<0.001, respectively), and significantly downregulated the expression levels of matrix metalloproteinase-2 (42±2.3%, P<0.05; 71±2.5%, P<0.01; and 95.8±3.3%, P<0.001, respectively) and aquaporin 4 (38±2.5%, P<0.05; 69±2.6%, P<0.01; and 88±3.0%, P<0.001, respectively), compared with the untreated cells. The wound healing assay demonstrated that cell migration was significantly impaired by treatment with 0.05, 0.5 and 5 µM pentamidine compared with untreated cells (88±4.2%, P<0.05; 64±2%, P<0.01; and 42±3.1%, P<0.001, respectively). Although additional in vivo studies are required to clarify the current in vitro data, the present study indicates that pentamidine and S100B-p53 inhibitors may represent a novel approach for the treatment of glioma.

15.
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
16.
Gut ; 63(8): 1300-12, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24082036

ABSTRACT

OBJECTIVE: Enteric glia activation has been reported to amplify intestinal inflammation via the enteroglial-specific S100B protein. This neurotrophin promotes macrophage recruitment in the mucosa, amplify colonic inflammation and interacts with toll-like receptors (TLR). Molecules inhibiting S100B-driven enteric activation might mitigate the course of ulcerative colitis (UC). This study aims to investigate the effects of palmitoylethanolammide (PEA), a drug able to counteract astroglial activation in the central nervous system, on intestinal inflammation, in humans and mice. DESIGN: Mouse models of dextran sodium sulphate (DSS)-induced colitis, colonic biopsies deriving from UC patients and primary cultures of mouse and human enteric glial cells (EGC), have been used to assess the effects of PEA, alone or in the presence of specific PPARα or PPARγ antagonists, on: macroscopic signs of UC (DAI score, colon length, spleen weight, macrophages/neutrophils infiltration); the expression and release of proinflammatory markers typical of UC; TLR pathway in EGCs. RESULTS: PEA treatment improves all macroscopic signs of UC and decreases the expression and release of all the proinflammatory markers tested. PEA anti-inflammatory effects are mediated by the selective targeting of the S100B/TLR4 axis on ECG, causing a downstream inhibition of nuclear factor kappa B (NF-kB)-dependent inflammation. Antagonists at PPARα, but not PPARγ, abolished PEA effects, in mice and in humans. CONCLUSIONS: Because of its lack of toxicity, its ability in reducing inflammation and its selective PPARα action, PEA might be an innovative molecule to broaden pharmacological strategies against UC.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colitis, Ulcerative/pathology , Endocannabinoids/therapeutic use , Ethanolamines/therapeutic use , Neuroglia/metabolism , PPAR alpha/metabolism , Palmitic Acids/therapeutic use , S100 Calcium Binding Protein beta Subunit/metabolism , Toll-Like Receptor 4/metabolism , Amides , Anilides/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cells, Cultured , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colon, Sigmoid/chemistry , Colon, Sigmoid/pathology , Cyclooxygenase 2/metabolism , Dextran Sulfate , Dinoprostone/metabolism , Endocannabinoids/pharmacology , Ethanolamines/pharmacology , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Indoles/pharmacology , Male , Mice , Middle Aged , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Neuroglia/drug effects , Neutrophil Infiltration/drug effects , Nitric Oxide/analysis , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , PPAR alpha/antagonists & inhibitors , PPAR gamma/antagonists & inhibitors , Palmitic Acids/pharmacology , Rectum/chemistry , Rectum/pathology , Severity of Illness Index , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
17.
Phytother Res ; 27(5): 633-6, 2013 May.
Article in English | MEDLINE | ID: mdl-22815234

ABSTRACT

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.


Subject(s)
Cannabidiol/therapeutic use , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Humans , Inflammation/drug therapy , Phytotherapy , Plant Extracts/therapeutic use
18.
J Neuroinflammation ; 9: 277, 2012 Dec 23.
Article in English | MEDLINE | ID: mdl-23259641

ABSTRACT

BACKGROUND: Intestinal inflammation is partly driven by enteroglial-derived S100B protein. The antiprotozoal drug pentamidine directly blocks S100B activity. We aimed to investigate the effect of pentamidine on intestinal inflammation using an animal model of dextran sodium sulphate (DSS)-induced acute colitis. METHODS: Mice were divided into: control group, colitis group (4% DSS for four days) and two pentamidine-treated colitis groups (0.8 mg/kg and 4 mg/kg). Anti-inflammatory effect of pentamidine was assessed in colonic tissue by evaluating the disease activity index and the severity of histological changes. Colonic tissue were also used to evaluate cyclooxigenase-2, inducible nitric oxide synthase, S100B, glial fibrillary acidic protein, phosphorylated-p38 MAPkinase, p50, p65 protein expression, malondyaldheyde production, mieloperoxidase activity, and macrophage infiltration. Nitric oxide, prostaglandin E2, interleukin-1 beta, tumor necrosis factor alpha, and S100B levels were detected in plasma samples. Parallel measurements were performed in vitro on dissected mucosa and longitudinal muscle myenteric plexus (LMMP) preparations after challenge with LPS + DSS or exogenous S100B protein in the presence or absence of pentamidine. RESULTS: Pentamidine treatment significantly ameliorated the severity of acute colitis in mice, as showed by macroscopic evaluation and histological/biochemical assays in colonic tissues and in plasma. Pentamidine effect on inflammatory mediators was almost completely abrogated in dissected mucosa but not in LMMP. CONCLUSIONS: Pentamidine exerts a marked anti-inflammatory effect in a mice model of acute colitis, likely targeting S100B activity. Pentamidine might be an innovative molecule to broaden pharmacological tools against colitis.


Subject(s)
Antiprotozoal Agents/therapeutic use , Colitis/drug therapy , Pentamidine/therapeutic use , Animals , Colitis/blood , Colitis/chemically induced , Colitis/pathology , Colon/metabolism , Colon/pathology , Cyclooxygenase 2/metabolism , Dextran Sulfate/toxicity , Dinoprostone/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Gene Expression Regulation/drug effects , Interleukin-1beta/blood , Macrophages/drug effects , Male , Mice , Mucous Membrane/pathology , Nerve Tissue Proteins/metabolism , Neutrophil Infiltration/drug effects , Nitrites/blood , Peroxidase/metabolism , Severity of Illness Index , Time Factors , Tumor Necrosis Factor-alpha/blood , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...