Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Curr Top Behav Neurosci ; 60: 73-87, 2023.
Article in English | MEDLINE | ID: mdl-35538302

ABSTRACT

Structural neuroplasticity in the adult brain is a process involving quantitative changes of the number and size of neurons and of their dendritic arborization, axon branching, spines, and synapses. These changes can occur in specific neural circuits as adaptive response to environmental challenges, exposure to stressors, tissue damage or degeneration. Converging studies point to evidence of structural plasticity in circuits operated by glutamate, GABA, dopamine, and serotonin neurotransmitters, in concert with neurotrophic factors such as Brain Derived Neurotrophic Factor (BDNF) or Insulin Growth Factor 1 (IGF1) and a series of modulators that include circulating hormones. Intriguingly, most of these endogenous agents trigger the activation of the PI3K/Akt/mTOR and ERK1/2 intracellular pathways that, in turn, lead to the production of growth-related structural changes, enhancing protein synthesis, metabolic enzyme functions, mitogenesis for energy, and new lipid-bilayer membrane apposition. The dopamine (DA) D3 receptor has been shown to play a specific role by inducing structural plasticity of the DAergic neurons of the nigrostriatal and mesocorticolimbic circuit, where they are expressed in rodents and humans, via activation of the mTORC1 and ERK1/2 pathways. These effects are BDNF-dependent and require the recruitment of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors to allow the structural changes. Since in mood disorders, depression and anhedonia have been proposed to be associated with impaired neuroplasticity and reduced DAergic tone in brain circuits connecting prefrontal cortex, ventral striatum, amygdala, and ventral mesencephalon, activation of D3 receptors could provide a therapeutic benefit. Sustained improvements of mood and anhedonia were observed in subjects with an unsatisfactory response to serotonin uptake inhibitors (SSRI) when treated with D3-preferential D2/D3 agonists such as pramipexole and ropinirole. The recent evidence that downstream mTOR pathway activation in human mesencephalic DA neurons is also produced by ketamine, probably the most effective antidepressant currently used in subjects with treatment-resistant depression, further supports the rationale for a D3 receptor activation in mood disorders.


Subject(s)
Brain-Derived Neurotrophic Factor , Receptors, Dopamine D3 , Humans , Receptors, Dopamine D3/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Dopamine , Anhedonia , Depression , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/pharmacology , Brain/metabolism , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/pharmacology , Dopaminergic Neurons/metabolism , Neuronal Plasticity
2.
Nutrition ; 106: 111895, 2023 02.
Article in English | MEDLINE | ID: mdl-36435090

ABSTRACT

Bowel obstruction and nutrient deficiencies are commonly encountered complications seen during follow-up of patients with short bowel syndrome. It is rare to see vitamin B12 deficiency among micronutrient deficits. The onset of B12 deficiency can be insidious, with autonomic features preceding other symptoms. In this study, we report a case in which vitamin B12 deficiency presented with intestinal dysmotility mimicking mechanical bowel obstruction.


Subject(s)
Intestinal Obstruction , Intestinal Pseudo-Obstruction , Short Bowel Syndrome , Vitamin B 12 Deficiency , Humans , Vitamin B 12 , Vitamin B 12 Deficiency/complications , Vitamin B 12 Deficiency/diagnosis , Short Bowel Syndrome/complications , Intestinal Pseudo-Obstruction/etiology , Intestinal Pseudo-Obstruction/complications , Vitamins
3.
Front Pharmacol ; 13: 913210, 2022.
Article in English | MEDLINE | ID: mdl-35721218

ABSTRACT

The epigenetic agents, L-acetylcarnitine (LAC) and L-methylfolate (MF) are putative candidates as add-on drugs in depression. We evaluated the effect of a combined treatment with LAC and MF in two different paradigms of chronic stress in mice and in human inducible pluripotent stem cells (hiPSCs) differentiated into dopaminergic neurons. Two groups of mice were exposed to chronic unpredictable stress (CUS) for 28 days or chronic restraint stress (CRS) for 21 day, and LAC (30 or 100 mg/kg) and/or MF (0.75 or 3 mg/kg) were administered i.p. once a day for 14 days, starting from the last week of stress. In both stress paradigms, LAC and MF acted synergistically in reducing the immobility time in the forced swim test and enhancing BDNF protein levels in the frontal cortex and hippocampus. In addition, LAC and MF acted synergistically in enhancing type-2 metabotropic glutamate receptor (mGlu2) protein levels in the hippocampus of mice exposed to CRS. Interestingly, CRS mice treated with MF showed an up-regulation of NFκB p65, which is a substrate for LAC-induced acetylation. We could also demonstrate a synergism between LAC and MF in cultured hiPSCs differentiated into dopamine neurons, by measuring dendrite length and number, and area of the cell soma after 3 days of drug exposure. These findings support the combined use of LAC and MF in the treatment of MDD and other stress-related disorders.

4.
Eur J Pharmacol ; 890: 173635, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33065094

ABSTRACT

F17464 (N-(3-{4-[4-(8-Oxo-8H-[1,3]-dioxolo-[4,5-g]-chromen-7-yl)-butyl]-piperazin-1-yl}-phenyl)-methanesulfonamide, hydrochloride) is a new potential antipsychotic with a unique profile. The compound exhibits high affinity for the human dopamine receptor subtype 3 (hD3) (Ki = 0.17 nM) and the serotonin receptor subtype 1a (5-HT1a) (Ki = 0.16 nM) and a >50 fold lower affinity for the human dopamine receptor subtype 2 short and long form (hD2s/l) (Ki = 8.9 and 12.1 nM, respectively). [14C]F17464 dynamic studies show a slower dissociation rate from hD3 receptor (t1/2 = 110 min) than from hD2s receptor (t1/2 = 1.4 min) and functional studies demonstrate that F17464 is a D3 receptor antagonist, 5-HT1a receptor partial agonist. In human dopaminergic neurons F17464 blocks ketamine induced morphological changes, an effect D3 receptor mediated. In vivo F17464 target engagement of both D2 and 5-HT1a receptors is demonstrated in displacement studies in the mouse brain. F17464 increases dopamine release in the rat prefrontal cortex and mouse lateral forebrain - dorsal striatum and seems to reduce the effect of MK801 on % c-fos mRNA medium expressing neurons in cortical and subcortical regions. F17464 also rescues valproate induced impairment in a rat social interaction model of autism. All the neurochemistry and behavioural effects of F17464 are observed in the dose range 0.32-2.5 mg/kg i.p. in both rats and mice. The in vitro - in vivo pharmacology profile of F17464 in preclinical models is discussed in support of a therapeutic use of the compound in schizophrenia and autism.


Subject(s)
Antipsychotic Agents/pharmacology , Benzopyrans/pharmacology , Dopamine Antagonists/pharmacology , Piperazines/pharmacology , Receptors, Dopamine D3/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Antipsychotic Agents/therapeutic use , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Behavior, Animal/drug effects , Benzopyrans/therapeutic use , Biogenic Monoamines/metabolism , Brain/drug effects , Brain/metabolism , Catalepsy/drug therapy , Cells, Cultured , Dopamine/metabolism , Dopamine Antagonists/therapeutic use , Dopaminergic Neurons/drug effects , Female , Genes, fos/drug effects , Male , Mice , Neuronal Plasticity/drug effects , Piperazines/therapeutic use , Prolactin/blood , Rats, Sprague-Dawley , Receptors, Dopamine D3/metabolism , Sulfonamides/therapeutic use , Valproic Acid/toxicity
5.
Chronic Stress (Thousand Oaks) ; 3: 2470547019842545, 2019.
Article in English | MEDLINE | ID: mdl-32440593

ABSTRACT

The mechanisms underlying the antidepressant effects of ketamine in treatment-resistant depression are only partially understood. Reactivation of neural plasticity in prefrontal cortex has been considered critical in mediating the effects of standard antidepressants, but in treatment-resistant depression patients with severe anhedonia, other components of the affected brain circuits, for example, the dopamine system, could be involved. In a recent article in Molecular Psychiatry, we showed that ketamine induces neural plasticity in human and mouse dopaminergic neurons. Human dopaminergic neurons were differentiated from inducible pluripotent stem cells for over 60 days. Mimicking the pharmacokinetic exposures occurring in treatment-resistant depression subjects, cultures were incubated with either ketamine at 0.1 and 1 µM for 1 h or with its active metabolite (2R,6R)-hydroxynorketamine at 0.1 and 0.5 µM for up to 6 h. Three days after dosing, we observed a concentration-dependent increase in dendritic arborization and soma size. These effects were mediated by the activation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor that triggered the pathways of mammalian target of rapamycin and extracellular signal-regulated kinase via the engagement of brain-derived neurotrophic factor signaling, as previously described in rodent prefrontal cortex. Interestingly, we found that neural plasticity induced by ketamine requires functionally intact dopamine D3 receptors. These data are in keeping with our recent observation that plasticity can be induced in human dopaminergic neurons by the D3 receptor-preferential agonist pramipexole, whose effect as augmentation treatment in treatment-resistant depression has been reported. Overall, the evidence of pharmacologic response in human inducible pluripotent stem cell-derived neurons could provide complementary information to those provided by circuit-based imaging when assessing the potential response to a given augmentation treatment.

6.
Neuroreport ; 30(3): 207-212, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30586092

ABSTRACT

The mechanisms underlying the prolonged antidepressant effects after a single exposure to ketamine are only partially understood. Converging findings indicate a critical role of structural neuroplasticity, recently also proposed for dopaminergic (DA) neurons known to be involved in a depression core symptom, anhedonia. We recently showed that ketamine induces dendritic outgrowth in human DA neurons differentiated in-vitro from induced pluripotent stem cells of healthy donors, a phenomenon blocked by the α-amino-3-hydroxy-5-methy-4-isoxazole propionate receptor antagonist NBQX. As changes in the expression of AMPA receptor subunits GluR1 and GluR2 were observed in neuroplasticity of rodent DA neurons, we aimed to explore this phenomenon in human DA neurons. Using specific antibodies against GluR1 and GluR2 α-amino-3-hydroxy-5-methy-4-isoxazole propionate receptor subunits, we showed that GluR1 levels were significantly higher in soma than in dendrites, whereas for GluR2, levels were significantly higher in dendrites than in soma. One hour exposure to 1 µM ketamine increased the signal of both subunits in dendrites, but only of GluR2 in soma, at 24, 48, and 72 h. Nonlinear polynomial fitting of dendritic expression indicated that the two curves were significantly different, with stronger and more sustained effects on GluR2 expression. Overall, these data support a role for GluR1 and GluR2 dendritic upregulation in driving structural plasticity in human DA neurons depending on ketamine transient exposure, indicating translationally relevant downstream mechanism possibly involved in antidepressant effects.


Subject(s)
Cell Differentiation/drug effects , Dopaminergic Neurons/drug effects , Induced Pluripotent Stem Cells/drug effects , Ketamine/pharmacology , Dendrites/metabolism , Dopaminergic Neurons/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Neuronal Plasticity/drug effects , Receptors, AMPA/drug effects
7.
Neuroreport ; 29(16): 1425-1430, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30211770

ABSTRACT

The mechanisms underlying the prolonged antidepressant effects after a single infusion of ketamine are only partially understood. Ketamine's half-life of ∼2 h cannot explain antidepressant effects that last for 1 week, suggesting the triggering of long-lasting neuroplasticity. Recent human pharmacokinetics (PK) data indicate that a ketamine metabolite, (2R,6R)-hydroxynorketamine (HNK), persists in the high submicromolar range for additional 6-12 h. As in rodents HNK can induce dendrite outgrowth through AMPA receptor-mediated mechanisms, in this work, we aimed to show that HNK produces similar effects in human neurons at concentrations and exposure time compatible with human PK after ketamine infusion. Human dopaminergic neurons were differentiated in-vitro from inducible pluripotent stem cells obtained from healthy donors. Exposure to submicromolar HNK for 6 h produced dendrite outgrowth when measured 3 days after exposure. This neuroplasticity was similar to that obtained with exposure to low micromolar concentrations of ketamine for 1 or 6 h. HNK and ketamine effects were blocked by pretreatment with the AMPA receptor antagonists NBQX and GYKI 52466, and by the mammalian target of rapamycin pathway blocker rapamycin. It is reasonable to conclude that the mechanistic similarity between ketamine and HNK and their diachronic brain exposure owing to the different plasma PK observed after single therapeutic ketamine infusion should contribute to the final sustained antidepressant action.


Subject(s)
Dendrites/drug effects , Induced Pluripotent Stem Cells/drug effects , Ketamine/pharmacology , Neurons/cytology , Neurons/drug effects , Receptors, AMPA/metabolism , Cells, Cultured , Dopamine/metabolism , Excitatory Amino Acid Agents/pharmacology , Gene Expression Regulation/drug effects , Humans , RNA, Messenger , Time Factors
8.
Front Pharmacol ; 9: 572, 2018.
Article in English | MEDLINE | ID: mdl-29910731

ABSTRACT

Midbrain dopamine (DA) neurons are considered a critical substrate for the reinforcing and sensitizing effects of nicotine and tobacco dependence. While the role of the α4 and ß2 subunit containing nicotinic acetylcholine receptors (α4ß2∗nAChRs) in mediating nicotine effects on DA release and DA neuron activity has been widely explored, less information is available on their role in the morphological adaptation of the DA system to nicotine, eventually leading to dysfunctional behaviors observed in nicotine dependence. In particular, no information is available on the role of α6∗nAChRs in nicotine-induced structural plasticity in rodents and no direct evidence exists regarding the occurrence of structural plasticity in human DA neurons exposed to nicotine. To approach this problem, we used two parallel in vitro systems, mouse primary DA neuron cultures from E12.5 embryos and human DA neurons differentiated from induced pluripotent stem cells (iPSCs) of healthy donors, identified using TH+ immunoreactivity. In both systems, nicotine 1-10 µM produced a dose-dependent increase of maximal dendrite length, number of primary dendrites, and soma size when measured after 3 days in culture. These effects were blocked by pretreatments with the α6∗nAChR antagonists α-conotoxin MII and α-conotoxin PIA, as well as by the α4ß2nAChR antagonist dihydro-ß-erythroidine (DHßE) in both mouse and human DA neurons. Nicotine was also ineffective when the primary DA neurons were obtained from null mutant mice for either the α6 subunit or both the α4 and α6 subunits of nAChR. When pregnant mice were exposed to nicotine from gestational day 15, structural plasticity was also observed in the midbrain DA neurons of postnatal day 1 offspring only in wild-type mice and not in both null mutant mice. This study confirmed the critical role of α4α6∗nAChRs in mediating nicotine-induced structural plasticity in both mouse and human DA neurons, supporting the translational relevance of neurons differentiated from human iPSCs for pharmacological studies.

9.
Neural Plast ; 2018: 4196961, 2018.
Article in English | MEDLINE | ID: mdl-29531524

ABSTRACT

The antiparkinsonian ropinirole and pramipexole are D3 receptor- (D3R-) preferring dopaminergic (DA) agonists used as adjunctive therapeutics for the treatment resistant depression (TRD). While the exact antidepressant mechanism of action remains uncertain, a role for D3R in the restoration of impaired neuroplasticity occurring in TRD has been proposed. Since D3R agonists are highly expressed on DA neurons in humans, we studied the effect of ropinirole and pramipexole on structural plasticity using a translational model of human-inducible pluripotent stem cells (hiPSCs). Two hiPSC clones from healthy donors were differentiated into midbrain DA neurons. Ropinirole and pramipexole produced dose-dependent increases of dendritic arborization and soma size after 3 days of culture, effects antagonized by the selective D3R antagonists SB277011-A and S33084 and by the mTOR pathway kinase inhibitors LY294002 and rapamycin. All treatments were also effective in attenuating the D3R-dependent increase of p70S6-kinase phosphorylation. Immunoneutralisation of BDNF, inhibition of TrkB receptors, and blockade of MEK-ERK signaling likewise prevented ropinirole-induced structural plasticity, suggesting a critical interaction between BDNF and D3R signaling pathways. The highly similar profiles of data acquired with DA neurons derived from two hiPSC clones underpin their reliability for characterization of pharmacological agents acting via dopaminergic mechanisms.


Subject(s)
Antiparkinson Agents/administration & dosage , Benzothiazoles/administration & dosage , Brain-Derived Neurotrophic Factor/metabolism , Dopaminergic Neurons , Indoles/administration & dosage , Neuronal Plasticity/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Dopaminergic Neurons/cytology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Humans , Induced Pluripotent Stem Cells/physiology , Mice , Pramipexole , Signal Transduction
10.
Neuroreport ; 29(8): 615-620, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29570499

ABSTRACT

Ketamine is a noncompetitive glutamate N-methyl-D-aspartic acid receptor antagonist. When acutely administered to rodents, it produces a rapid antidepressant effect. There is evidence that N-methyl-D-aspartic acid receptor blockade enhances glutamatergic transmission preferentially engaging α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors leading to mTOR (mammalian target of rapamycin) pathways activation, thus resulting into downstream neuroadaptive changes in limbic structures. Recent in-vitro data on primary neuronal cultures showed that ketamine activates mTOR also in dopaminergic neurons, and this activation depends on the presence of functional dopamine D3 receptors. The aim of this work was to study the in-vivo relevance of viable D3 receptors in mediating the effects of acute ketamine administration on the mTOR downstream substrate p70 ribosomal S6 kinase (p70S6K), an obligatory substrate for mTOR. We compared the effects of single ketamine 5 mg/kg, 10 mg/kg, or vehicle injection in wild-type and D3 receptor knockout mice. Animals were killed after 60 min, and their brains were processed for p-p70S6K immunohistochemistry. Ketamine increased p70S6K phosphorylation in prefrontal cortex, nucleus accumbens core and shell, ventral tegmental area, substantia nigra, hippocampal CA1, CA2, and CA3, and basolateral amygdala of wild-type mice but not in mutant mice. Our study demonstrates that ketamine-induced p70S6K phosphorylation is dependent on viable D3R expressed in most of limbic structures.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Limbic System/drug effects , Receptors, Dopamine D3/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Limbic System/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Dopamine D3/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
11.
Hemoglobin ; 41(4-6): 314-316, 2017.
Article in English | MEDLINE | ID: mdl-29313431

ABSTRACT

Approximately 150 variants described in the HbVar database have been found to be unstable and about 80.0% of these are on the ß-globin gene. We describe the case of a 3-year-old child who presented at the emergency room with fever and asthenia. Hematological data suggested severe hemolytic anemia. Sequencing of the ß-globin gene revealed the mutation HBB: c.278A>G at codon 92 in a heterozygous state, reported as Hb Mozhaisk in the HbVar database. Other family members did not have Hb Mozhaisk, thus, this variant is due to a de novo mutation. Because of the rarity of this globin variant, we believe it is important to report similar cases, to have a more complete phenotype description of the pathology and define an adequate reproductive risk for couples, considering the dominant inheritance pattern (hence an inheritance risk of 50.0%).


Subject(s)
Anemia, Hemolytic/genetics , Codon , Hemoglobins, Abnormal/genetics , Point Mutation , Child, Preschool , Humans , Male
12.
Pediatr Hematol Oncol ; 32(3): 223-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25551550

ABSTRACT

Neuroblastoma presenting with obstructive jaundice is a rare event. Management of this condition includes surgery, chemotherapy, radiotherapy, temporary cholecystostomy tube, endoscopic retrograde cholangiopancreatography (ERCP), and internal biliary drainage (IBD). We herein describe our experience with one infant affected by neuroblastoma presenting with jaundice, who successfully underwent percutaneous transhepatic biliary drainage (PTBD). This report introduces PTBD as a viable treatment option for neuroblastoma and obstructive jaundice and provides a review of the pertinent literature.


Subject(s)
Cholestasis/surgery , Drainage/methods , Jaundice, Obstructive/surgery , Neuroblastoma/complications , Retroperitoneal Neoplasms/complications , Cholangiopancreatography, Endoscopic Retrograde , Humans , Infant , Jaundice, Obstructive/etiology , Male
13.
Front Pharmacol ; 5: 259, 2014.
Article in English | MEDLINE | ID: mdl-25505416

ABSTRACT

Mesencephalic dopaminergic neurons were suggested to be a critical physiopathology substrate for addiction disorders. Among neuroadaptive processes to addictive drugs, structural plasticity has attracted attention. While structural plasticity occurs at both pre- and post-synaptic levels in the mesolimbic dopaminergic system, the present review focuses only on dopaminergic neurons. Exposures to addictive drugs determine two opposite structural responses, hypothrophic plasticity produced by opioids and cannabinoids (in particular during the early withdrawal phase) and hypertrophic plasticity, mostly driven by psychostimulants and nicotine. In vitro and in vivo studies identified BDNF and extracellular dopamine as two critical factors in determining structural plasticity, the two molecules sharing similar intracellular pathways involved in cell soma and dendrite growth, the MEK-ERK1/2 and the PI3K-Akt-mTOR, via preferential activation of TrkB and dopamine D3 receptors, respectively. At present information regarding specific structural changes associated to the various stages of the addiction cycle is incomplete. Encouraging neuroimaging data in humans indirectly support the preclinical evidence of hypotrophic and hypertrophic effects, suggesting a possible differential engagement of dopamine neurons in parallel and partially converging circuits controlling motivation, stress, and emotions.

14.
Pediatr Blood Cancer ; 61(9): 1593-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24757164

ABSTRACT

BACKGROUND: The aim of this study was to assess the prognostic value of multifocality and the effectiveness of two different therapeutic strategies in patients with newly diagnosed hepatoblastoma. PROCEDURES: Between 1998 and 2011, 31 patients diagnosed with hepatoblastoma were referred to Ospedale Papa Giovanni XXIII, Bergamo, Italy. Patients were stratified according to SIOPEL protocols into high-risk (HR if AFP <100 ng/mL and/or PRETEXT IV and/or vascular invasion and/or extra-hepatic intra-abdominal disease and/or metastases) and standard-risk (SR, all others). The patient data we evaluated were: multifocality; patient age; gender; platelet count; AFP level at diagnosis, during treatment and follow-up; histotype; gestational age; birth weight; surgery (either resection or transplantation) and chemotherapy regimen adopted before and after surgery. The outcome measures were event free survival (EFS) and overall survival (OS); survival curves were estimated according to Kaplan-Meier. RESULTS: EFS and OS were associated significantly with multifocality (3-year EFS 40% vs. 95%, P = 0.006; 3-year OS 42% vs. 95%, P = 0.004). Multivariate analysis demonstrated that multifocality predicts lower EFS (hazard ratio 10.01, P = 0.007). Other factors at diagnosis did not reach statistical significance. A marked treatment dependent improvement was associated with intensive chemotherapy given both before and after liver transplantation (P = 0.06). CONCLUSIONS: Patients diagnosed with multifocal tumors had lower EFS levels. Multifocality should be taken into account for future stratification and further studied to assess genetic profile, immunochemistry and prognostic role.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hepatoblastoma/secondary , Liver Neoplasms/pathology , Child , Child, Preschool , Combined Modality Therapy , Female , Follow-Up Studies , Hepatoblastoma/mortality , Hepatoblastoma/therapy , Humans , Infant , Infant, Newborn , Liver Neoplasms/mortality , Liver Neoplasms/therapy , Male , Multivariate Analysis , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Retrospective Studies , Survival Rate
15.
Ital J Pediatr ; 39: 65, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24148885

ABSTRACT

We report the use of high dose chemotherapy with peripheral blood stem cell rescue as a consolidation treatment for a 3-year-old child affected by metastatic hepatoblastoma, who achieved complete lung response only after conventional treatment. The patient is presently alive 27 months after high dose chemotherapy with blood stem cell rescue with no evidence of disease.The role of high dose chemotherapy with blood stem cell rescue to consolidate the complete clearing of lung disease in metastatic hepatoblastoma remains controversial; the data available in the literature and our experience seems to suggest to keep this treatment option open to further consideration in the clinical setting of high-risk patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Hepatoblastoma/secondary , Hepatoblastoma/therapy , Liver Neoplasms/therapy , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Biopsy, Needle , Child, Preschool , Combined Modality Therapy , Dose-Response Relationship, Drug , Follow-Up Studies , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Neoplasm Invasiveness/pathology , Neoplasm Staging , Risk Assessment , Time Factors , Tomography, X-Ray Computed/methods , Treatment Outcome
16.
Mol Pharmacol ; 83(6): 1176-89, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23543412

ABSTRACT

Although long-term exposure to nicotine is highly addictive, one beneficial consequence of chronic tobacco use is a reduced risk for Parkinson's disease. Of interest, these effects both reflect structural and functional plasticity of brain circuits controlling reward and motor behavior and, specifically, recruitment of nicotinic acetylcholine receptors (nAChR) in mesencephalic dopaminergic neurons. Because the underlying cellular mechanisms are poorly understood, we addressed this issue with use of primary cultures of mouse mesencephalic dopaminergic neurons. Exposure to nicotine (1-10 µM) for 72 hours in vitro increased dendritic arborization and soma size in primary cultures. These effects were blocked by mecamylamine and dihydro-ß-erythroidine, but not methyllycaconitine. The involvement of α4ß2 nAChR was supported by the lack of nicotine-induced structural remodeling in neurons from α4 null mutant mice (KO). Challenge with nicotine triggered phosphorylation of the extracellular signal-regulated kinase (ERK) and the thymoma viral proto-oncogene (Akt), followed by activation of the mammalian target of rapamycin complex 1 (mTORC1)-dependent p70 ribosomal S6 protein kinase. Upstream pathway blockade using the phosphatidylinositol 3-kinase inhibitor LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one hydrochloride] resulted in suppression of nicotine-induced phosphorylations and structural plasticity. These effects were dependent on functional DA D3 receptor (D3R), because nicotine was inactive both in cultures from D3R KO mice and after pharmacologic blockade with D3R antagonist trans-N-4-2-(6-cyano-1,2,3, 4-tetrahydroisoquinolin-2-yl)ethylcyclohexyl-4-quinolinecarboxamide (SB-277011-A) (50 nM). Finally, exposure to nicotine in utero (5 mg/kg/day for 5 days) resulted in increased soma area of DAergic neurons of newborn mice, effects not observed in D3 receptor null mutant mice mice. These findings indicate that nicotine-induced structural plasticity at mesencephalic dopaminergic neurons involves α4ß2 nAChRs together with dopamine D3R-mediated recruitment of ERK/Akt-mTORC1 signaling.


Subject(s)
Dopaminergic Neurons/drug effects , Mesencephalon/drug effects , Nicotine/pharmacology , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Dopamine D3/metabolism , Animals , Animals, Newborn , Cells, Cultured , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Enzyme Activation , Female , MAP Kinase Signaling System , Maternal Exposure , Maternal-Fetal Exchange , Mechanistic Target of Rapamycin Complex 1 , Mesencephalon/cytology , Mesencephalon/metabolism , Mice , Mice, Knockout , Multiprotein Complexes , Pregnancy , Receptors, Dopamine D3/genetics , Receptors, Nicotinic/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
17.
J Neurochem ; 120(5): 765-78, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22145570

ABSTRACT

Exposure to psychostimulants results in neuroadaptive changes of the mesencephalic dopaminergic system including morphological reorganization of dopaminergic neurons. Increased dendrite arborization and soma area were previously observed in primary cultures of mesencephalic dopaminergic neurons after 3-day exposure to dopamine agonists via activation of D(3) autoreceptors (D(3) R). In this work, we showed that cocaine significantly increased dendritic arborization and soma area of dopaminergic neurons from E12.5 mouse embryos by activating phosphorylation of extracellular signal-regulated kinase (ERK) and thymoma viral proto-oncogene (Akt). These effects were dependent on functional D(3) R expression because cocaine did not produce morphological changes or ERK/Akt phosphorylation neither in primary cultures of D(3) R mutant mice nor following pharmacologic blockade with D(3) R antagonists SB-277011-A and S-33084. Cocaine effects on morphology and ERK/Akt phosphorylation were inhibited by pre-incubation with the phosphatidylinositol 3-kinase inhibitor LY294002. These observations were corroborated in vivo by morphometrical assessment of mesencephalic dopaminergic neurons of P1 newborns exposed to cocaine from E12.5 to E16.5. Cocaine increased the soma area of wild-type but not of D(3) R mutant mice, supporting the translational value of primary culture. These findings indicate a direct involvement of D3R and ERK/Akt pathways as critical mediators of cocaine-induced structural plasticity, suggesting their involvement in psychostimulant addiction.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopaminergic Neurons/drug effects , Mesencephalon/cytology , Neurons/physiology , Presynaptic Terminals/metabolism , Receptors, Dopamine D3/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Dopamine , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , In Vitro Techniques , Mice , Mice, Knockout , Neurons/drug effects , Oncogene Protein v-akt/metabolism , Presynaptic Terminals/drug effects , Receptors, Dopamine D3/deficiency , Signal Transduction/drug effects , Tritium/metabolism , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...