Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Rev Recent Clin Trials ; 12(3): 187-192, 2017.
Article in English | MEDLINE | ID: mdl-28814259

ABSTRACT

INTRODUCTION: 13C-urea breath test (UBT) is a non-invasive test for detecting active H. pylori infection. Previous studies showed a correlation of delta over baseline (DOB) values with bacterial load, mucosal inflammation and successful eradication. Gender has been shown to affect DOB in children. Aim of our study was to verify whether gender or ethnicity affects DOB in adults. PATIENTS AND METHODS: We retrospectively analyzed data of 2922 patients (1024M/1898F mean age 47±15 years) that underwent UBT in our outpatient unit, from October 2015 to October 2016. Patients were divided based on gender and ethnicity; mean DOB values were then compared. RESULTS: 686 pts (23.4%, 258M/428F, mean age 45±17 years) of 2922 pts showed a positive UBT. Prevalence of H. pylori infection was significantly higher in males compared to females (29% vs 22%; p=0,03). Females showed a significant higher mean DOB (34±25 vs 27,6±22; p=0,008). A total of 2922 UBT were performed during the study period (F:1898, 65%; M: 1024 35%). The prevalence of H. pylori infection is 32% in those from Eastern Countries, 28% in those from South America and 40% in both those coming from Africa and Asia. We found significantly lower DOB values in Italians compared to non-Italian (mean DOB 36±27 vs 69±32; p<0.0001). CONCLUSION: Our study showed that in our geographic area, prevalence H. pylori infection is higher in males. Moreover, it demonstrates for the first time in our geographic area that adult females show a significantly higher DOB compared to males (p=0,008). Whether this effect may be due to hormonal differences, able to influence gastric emptying, bacterial load, or even the production of urease by H. pylori, merits further investigation.


Subject(s)
Breath Tests/methods , Ethnicity , Helicobacter Infections/diagnosis , Helicobacter pylori/isolation & purification , Risk Assessment/methods , Urea/analysis , Female , Helicobacter Infections/ethnology , Helicobacter Infections/metabolism , Humans , Italy/epidemiology , Male , Middle Aged , Retrospective Studies , Sex Distribution , Sex Factors
2.
Dis Markers ; 2016: 5340386, 2016.
Article in English | MEDLINE | ID: mdl-28070137

ABSTRACT

Aim. Lactulose/mannitol ratio is used to assess intestinal barrier function. Aim of this work was to develop a robust and rapid method for the analysis of lactulose and mannitol in urine by liquid chromatography coupled to tandem mass spectrometry. Lactulose/mannitol ratio has been measured in pediatric patients suffering from irritable bowel syndrome. Methods. Calibration curves and raffinose, used as internal standard, were prepared in water : acetonitrile 20 : 80. Fifty µL of urine sample was added to 450 µL of internal standard solution. The chromatographic separation was performed using a Luna NH2 column operating at a flow rate of 200 µL/min and eluted with a linear gradient from 20% to 80% water in acetonitrile. Total run time is 9 minutes. The mass spectrometry operates in electrospray negative mode. Method was fully validated according to European Medicine Agency guidelines. Results and Conclusions. Linearity ranged from 10 to 1000 mg/L for mannitol and 2.5 to 1000 mg/L for lactulose. Imprecision in intra- and interassay was lower than 15% for both analytes. Accuracy was higher than 85%. Lactulose/mannitol ratio in pediatric patients is significantly higher than that measured in controls. The presented method, rapid and sensitive, is suitable in a clinical laboratory.


Subject(s)
Irritable Bowel Syndrome/urine , Lactulose/urine , Mannitol/urine , Urinalysis/methods , Adolescent , Case-Control Studies , Child , Chromatography, Liquid/methods , Female , Humans , Male , Mass Spectrometry/methods
3.
J Pathol ; 236(4): 479-90, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25875314

ABSTRACT

Chronic inflammation is a leading cause of neoplastic transformation in many human cancers and especially in colon cancer (CC), in part due to tumour promotion by nitric oxide (NO) generated at inflammatory sites. It has also been suggested that high NO synthesis, secondary to inducible NO synthase (iNOS) expression, is a distinctive feature of cancer stem cells (CSCs), a small subset of tumour cells with self-renewal capacity. In this study we explored the contribution of NO to the development of colon CSC features and evaluated potential strategies to treat CC by modulating NO production. Our data show an integral role for endogenous NO and iNOS activity in the biology of colon CSCs. Indeed, colon CSCs with high endogenous NO production (NO(high)) displayed higher tumourigenic abilities than NO(low) fractions. The blockade of endogenous NO availability, using either a specific iNOS inhibitor or a genetic knock-down of iNOS, resulted in a significant reduction of colon CSC tumourigenic capacities in vitro and in vivo. Interestingly, analysis of genes altered by iNOS-directed shRNA showed that the knockdown of iNOS expression was associated with a significant down-regulation of signalling pathways involved in stemness and tumour progression in colon CSCs. These findings confirm that endogenous NO plays an important role in defining the stemness properties of colon CSCs through cross-regulation of several cellular signalling pathways. This discovery could shed light on the mechanisms by which NO induces the growth and invasiveness of CC, providing new insights into the link between inflammation and colon tumourigenesis.


Subject(s)
Colorectal Neoplasms/enzymology , Neoplastic Stem Cells/enzymology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , AC133 Antigen , Animals , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Caco-2 Cells , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Peptides/metabolism , RNA Interference , Signal Transduction , Time Factors , Transfection , Tumor Burden , Up-Regulation , Xenograft Model Antitumor Assays
4.
Sci Rep ; 5: 9149, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25779766

ABSTRACT

Although the only effective drug against primary hepatocarcinoma, the multikinase inhibitor Sorafenib (SFB) usually fails to eradicate liver cancer. Since SFB targets mitochondria, cell metabolic reprogramming may underlie intrinsic tumor resistance. To characterize cancer cell metabolic response to SFB, we measured oxygen consumption, generation of reactive oxygen species (ROS) and ATP content in rat LCSC (Liver Cancer Stem Cells) -2 cells exposed to the drug. Genome wide analysis of gene expression was performed by Affymetrix technology. SFB cytotoxicity was evaluated by multiple assays in the presence or absence of metabolic inhibitors, or in cells genetically depleted of mitochondria. We found that low concentrations (2.5-5 µM) of SFB had a relatively modest effect on LCSC-2 or 293 T cell growth, but damaged mitochondria and increased intracellular ROS. Gene expression profiling of SFB-treated cells was consistent with a shift toward aerobic glycolysis and, accordingly, SFB cytotoxicity was dramatically increased by glucose withdrawal or the glycolytic inhibitor 2-DG. Under metabolic stress, activation of the AMP dependent Protein Kinase (AMPK), but not ROS blockade, protected cells from death. We conclude that mitochondrial damage and ROS drive cell killing by SFB, while glycolytic cell reprogramming may represent a resistance strategy potentially targetable by combination therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy , Cell Line, Tumor , Cell Respiration/drug effects , Cell Survival/drug effects , Deoxyglucose/pharmacology , Energy Metabolism/drug effects , Glycolysis/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Niacinamide/pharmacology , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sorafenib , TOR Serine-Threonine Kinases/metabolism
5.
Biomed Res Int ; 2014: 859871, 2014.
Article in English | MEDLINE | ID: mdl-24527460

ABSTRACT

There is an emerging body of evidence that chemoresistance and minimal residual disease result from selective resistance of a cell subpopulation from the original tumor that is molecularly and phenotypically distinct. These cells are called "cancer stem cells" (CSCs). In this review, we analyze the potential targeting strategies for eradicating CSCs specifically in order to develop more effective therapeutic strategies for metastatic colon cancer. These include induction of terminal epithelial differentiation of CSCs or targeting some genes expressed only in CSCs and involved in self-renewal and chemoresistance. Ideal targets could be cell regulators that simultaneously control the stemness and the resistance of CSCs. Another important aspect of cancer biology, which can also be harnessed to create novel broad-spectrum anticancer agents, is the Warburg effect, also known as aerobic glycolysis. Actually, little is yet known with regard to the metabolism of CSCs population, leaving an exciting unstudied avenue in the dawn of the emerging field of metabolomics.


Subject(s)
Colonic Neoplasms , Drug Delivery Systems , Drug Resistance, Neoplasm , Neoplastic Stem Cells , Animals , Antineoplastic Agents , Cell Line, Tumor , Humans , Mice
6.
World J Gastroenterol ; 19(29): 4671-8, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23922464

ABSTRACT

Among somatic stem cells, those residing in the intestine represent a fascinating and poorly explored research field. Particularly, somatic stem cells reside in the small intestine at the level of the crypt base, in a constant balance between self-renewal and differentiation. Aim of the present review is to delve into the mechanisms that regulate the delicate equilibrium through which intestinal stem cells orchestrate intestinal architecture. To this aim, special focus will be addressed to identify the integrating signals from the surrounding niche, supporting a model whereby distinct cell populations facilitate homeostatic vs injury-induced regeneration.


Subject(s)
Intestinal Diseases , Intestinal Mucosa , Intestines , Stem Cells , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Humans , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Intestinal Diseases/physiopathology , Intestinal Mucosa/metabolism , Intestines/pathology , Intestines/physiopathology , Regeneration , Signal Transduction , Stem Cell Niche , Stem Cells/metabolism , Stem Cells/pathology
7.
World J Gastroenterol ; 19(20): 2997-3006, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23716979

ABSTRACT

Tumors have long been viewed as a population in which all cells have the equal propensity to form new tumors, the so called conventional stochastic model. The cutting-edge theory on tumor origin and progression, tends to consider cancer as a stem cell disease. Stem cells are actively involved in the onset and maintenance of colon cancer. This review is intended to examine the state of the art on colon cancer stem cells (CSCs), with regard to the recent achievements of basic research and to the corresponding translational consequences. Specific prominence is given to the hypothesized origin of CSCs and to the methods for their identification. The growing understanding of CSC biology is driving the optimization of novel anti-cancer targeted drugs.


Subject(s)
Colonic Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Humans , Molecular Targeted Therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Signal Transduction
8.
Bone ; 52(1): 474-84, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22982077

ABSTRACT

Sporadic mono-sutural craniosynostosis represents a highly prevalent regional bone disorder, where a single cranial suture undergoes premature ossification due to a generally unclear etiopathogenesis. The LIM mineralization protein (LMP) was recently described as an efficient osteogenic molecule involved in osteoblast differentiation, expressed in calvarial tissues upon corticosteroid-osteogenic induction and used as a potent inducer of bone formation in several animal models. In this study, calvarial cells isolated from both prematurely fused and physiologically patent sutures of children with sporadic craniosynostosis, were used as an in vitro paradigmatic model for the study of the molecular events involved in calvarial osteogenesis, focusing on the possible role of the LMP-related osteogenic signaling. Calvarial cells isolated from both patent and fused sutures expressed a mesenchymal-like immunophenotype. Cells isolated from fused sutures displayed an increased osteogenic potential, being able to undergo spontaneous mineralization and premature response to osteogenic induction, leading to in vitro bone nodule formation. The expression of LMP and its target genes (bone morphogenetic protein-2, osteocalcin and Runt-related transcription factor 2) was significantly up-regulated in cells derived from the fused sutures. Upon silencing the expression of LMP in fused suture-derived cells, the osteogenic potential along with the expression of osteo-specific transcription factors decreased, restoring the "physiologic" cell behavior. These results suggested that: 1. mesenchymal cells residing in fused sutures display a constitutionally active osteogenic disposition leading to the premature suture ossification; 2. the molecular basis of the overactive osteogenic process may at least in part involve a deregulation of the LMP-related pathway in calvarial cells.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Craniosynostoses/physiopathology , Cytoskeletal Proteins/physiology , LIM Domain Proteins/physiology , Ossification, Heterotopic/physiopathology , Skull/physiopathology , Base Sequence , Child, Preschool , DNA Primers , Humans , Infant , Real-Time Polymerase Chain Reaction , Signal Transduction , Smad Proteins/metabolism
9.
J Biomed Biotechnol ; 2011: 860578, 2011.
Article in English | MEDLINE | ID: mdl-22228987

ABSTRACT

Mesenchymal stem cells (MSCs), represent an attractive tool for the establishment of a successful stem-cell-based therapy of liver diseases. A number of different mechanisms contribute to the therapeutic effects exerted by MSCs, since these cells can differentiate into functional hepatic cells and can also produce a series of growth factors and cytokines able to suppress inflammatory responses, reduce hepatocyte apoptosis, regress liver fibrosis, and enhance hepatocyte functionality. To date, the infusion of MSCs or MSC-conditioned medium has shown encouraging results in the treatment of fulminant hepatic failure and in end-stage liver disease in experimental settings. However, some issues under debate hamper the use of MSCs in clinical trials. This paper summarizes the biological relevance of MSCs and the potential benefits and risks that can result from translating the MSC research to the treatment of liver diseases.


Subject(s)
Liver Diseases/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Humans , Immune System/immunology , Liver Diseases/pathology , Liver Transplantation , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/immunology , Risk Factors
10.
Aging (Albany NY) ; 2(8): 487-503, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20739737

ABSTRACT

Deregulated nutrient signaling plays pivotal roles in body ageing and in diabetic complications; biochemical cascades linking energy dysmetabolism to cell damage and loss are still incompletely clarified, and novel molecular paradigms and pharmacological targets critically needed. We provide evidence that in the retrovirus-packaging cell line HEK293-T Phoenix, massive cell death in serum-free medium is remarkably prevented or attenuated by either glucose or aminoacid withdrawal, and by the glycolysis inhibitor 2-deoxy-glucose. A similar protection was also elicited by interference with mitochondrial function, clearly suggesting involvement of energy metabolism in increased cell survival. Oxidative stress did not account for nutrient toxicity on serum-starved cells. Instead, nutrient restriction was associated with reduced activity of the mTOR/S6 Kinase cascade. Moreover, pharmacological and genetic manipulation of the mTOR pathway modulated in an opposite fashion signaling to S6K/S6 and cell viability in nutrient-repleted medium. Additionally, stimulation of the AMP-activated Protein Kinase concomitantly inhibited mTOR signaling and cell death, while neither event was affected by overexpression of the NAD+ dependent deacetylase Sirt-1, another cellular sensor of nutrient scarcity. Finally, blockade of the mTOR cascade reduced hyperglycemic damage also in a more pathophysiologically relevant model, i.e. in human umbilical vein endothelial cells (HUVEC) exposed to hyperglycemia. Taken together these findings point to a key role of the mTOR/S6K cascade in cell damage by excess nutrients and scarcity of growth-factors, a condition shared by diabetes and other ageing-related pathologies.


Subject(s)
Cell Survival/physiology , Food Deprivation/physiology , Intracellular Signaling Peptides and Proteins/physiology , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/physiology , Antimetabolites/administration & dosage , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media, Serum-Free , Deoxyglucose/administration & dosage , HEK293 Cells , Humans , Mitochondria/physiology , Oxidative Stress/physiology , Ribosomal Protein S6/physiology , Ribosomal Protein S6 Kinases/physiology , Signal Transduction/physiology , Sirtuin 1/physiology , TOR Serine-Threonine Kinases/toxicity
11.
Proc Natl Acad Sci U S A ; 107(30): 13420-5, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20624962

ABSTRACT

Obesity and metabolic syndrome result from excess calorie intake and genetic predisposition and are mechanistically linked to type II diabetes and accelerated body aging; abnormal nutrient and insulin signaling participate in this pathologic process, yet the underlying molecular mechanisms are incompletely understood. Mice lacking the p66 kDa isoform of the Shc adaptor molecule live longer and are leaner than wild-type animals, suggesting that this molecule may have a role in metabolic derangement and premature senescence by overnutrition. We found that p66 deficiency exerts a modest but significant protective effect on fat accumulation and premature death in lepOb/Ob mice, an established genetic model of obesity and insulin resistance; strikingly, however, p66 inactivation improved glucose tolerance in these animals, without affecting (hyper)insulinaemia and independent of body weight. Protection from insulin resistance was cell autonomous, because isolated p66KO preadipocytes were relatively resistant to insulin desensitization by free fatty acids in vitro. Biochemical studies revealed that p66shc promotes the signal-inhibitory phosphorylation of the major insulin transducer IRS-1, by bridging IRS-1 and the mTOR effector p70S6 kinase, a molecule previously linked to obesity-induced insulin resistance. Importantly, IRS-1 was strongly up-regulated in the adipose tissue of p66KO lepOb/Ob mice, confirming that effects of p66 on tissue responsiveness to insulin are largely mediated by this molecule. Taken together, these findings identify p66shc as a major mediator of insulin resistance by excess nutrients, and by extension, as a potential molecular target against the spreading epidemic of obesity and type II diabetes.


Subject(s)
Insulin Resistance/physiology , Leptin/metabolism , Obesity/physiopathology , Shc Signaling Adaptor Proteins/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cells, Cultured , Flow Cytometry , Glucose Intolerance/genetics , Glucose Intolerance/physiopathology , Hyperinsulinism/genetics , Hyperinsulinism/physiopathology , Hypoglycemic Agents/pharmacology , Immunoblotting , Insulin/pharmacology , Leptin/genetics , Longevity/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Obese , Obesity/genetics , Obesity/metabolism , Phosphorylation , RNA Interference , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Shc Signaling Adaptor Proteins/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1
12.
J Hepatol ; 51(1): 77-92, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19446912

ABSTRACT

BACKGROUND/AIMS: Oval cells (OCs), putative hepatic stem cells, may give rise to liver cancers. We developed a carcinogenesis regimen, based upon induction of OC proliferation prior to carcinogen exposure. In our model, rats subjected to 2-acetylaminofluorene/ partial-hepatectomy followed by aflatoxin injection (APA regimen) developed well-differentiated hepatocholangiocarcinomas. The aim of this study was to establish and characterize cancer cell lines from this animal model. METHODS: Cancer cells were cultured from animals sacrificed eight months after treatment, and single clones were selected. The established cell lines, named LCSCs, were characterized, and their tumorigenicity was assessed in vivo. The roles of granulocyte-colony stimulating factor (G-CSF) and hepatocyte growth factor (HGF) in LCSC growth, survival and motility were also investigated. RESULTS: From primary tumors, six cell lines were developed. LCSCs shared with the primary tumors the expression of various OC-associated markers, including cMet and G-CSF receptor. In vitro, HGF conferred protection from death by serum withdrawal. Stimulation with G-CSF increased LCSC growth and motility, while the blockage of its receptor inhibited LCSC proliferation and migration. CONCLUSIONS: Six cancer cell lines were established from our model of hepatocholangiocarcinoma. HGF modulated LCSC resistance to apoptosis, while G-CSF acted on LCSCs as a proliferative and chemotactic agent.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cholangiocarcinoma/pathology , Granulocyte Colony-Stimulating Factor/physiology , Hepatocyte Growth Factor/physiology , Liver Neoplasms/pathology , 2-Acetylaminofluorene/toxicity , Aflatoxin B1/toxicity , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Hepatectomy , Male , Neoplasm Metastasis , Proto-Oncogene Proteins c-met/physiology , Rats , Rats, Inbred F344 , Receptors, Granulocyte Colony-Stimulating Factor/physiology
13.
Brain Res ; 1043(1-2): 32-41, 2005 May 10.
Article in English | MEDLINE | ID: mdl-15862515

ABSTRACT

Excessive activation of non-NMDA receptors, AMPA and kainate, contributes to neuronal degeneration in acute and progressive pathologies, possibly including schizophrenia. Because 5-HT(1A) receptor agonists have neuroprotective properties (e.g., against NMDA-induced neurotoxicity), we compared the effects of the antipsychotics, clozapine, ziprasidone and aripiprazole, that are partial agonists at 5-HT(1A) receptor, with those of haloperidol, which is devoid of 5-HT(1A) agonist properties, on kainic acid (KA)-induced striatal lesion volumes, in C57Bl/6N mice. The involvement of 5-HT(1A) receptors was determined by antagonist studies with WAY100635, and data were compared with those obtained using the potent and high efficacy 5-HT(1A) receptor agonist, F13714. Intra-striatal KA lesioning and measurement of lesion volumes using cresyl violet staining were carried out at 48 h after surgery. F13714, antipsychotics or vehicle were administered ip twice, 30 min before and 3 1/2 h after KA injection. WAY100635 (0.63 mg/kg) or vehicle were given sc 30 min before each drug injection. Clozapine (2 x 10 mg/kg), ziprasidone (2 x 20 mg/kg) and aripiprazole (2 x 10 mg/kg) decreased lesion volume by 61%, 59% and 73%, respectively. WAY100635 antagonized the effect of ziprasidone and of aripiprazole but only slightly attenuated that of clozapine. In contrast, haloperidol (2 x 0.16 mg/kg) did not affect KA-induced lesion volume. F13714 dose-dependently decreased lesion volume. The 61% decrease of lesion volume obtained with F13714 (2 x 0.63 mg/kg) was antagonized by WAY100635. WAY100635 alone did not affect lesion volume. These results show that 5-HT(1A) receptor activation protects against KA-induced striatal lesions and indicate that some atypical antipsychotic agents with 5-HT(1A) agonist properties may protect against excitotoxic injury, in vivo.


Subject(s)
Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Corpus Striatum/drug effects , Excitatory Amino Acid Agonists/toxicity , Kainic Acid/toxicity , Receptor, Serotonin, 5-HT1A/physiology , Aminopyridines/pharmacology , Animals , Aripiprazole , Corpus Striatum/pathology , Corpus Striatum/physiology , Disease Models, Animal , Haloperidol/pharmacology , Male , Mice , Mice, Inbred C57BL , Piperazines/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Quinolones/pharmacology , Schizophrenia/drug therapy , Schizophrenia/physiopathology , Serotonin 5-HT1 Receptor Agonists , Serotonin Antagonists/pharmacology , Thiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...