Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Exp Clin Cancer Res ; 43(1): 13, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38191418

ABSTRACT

BACKGROUND: Inflammation in the eye is often associated with aggravated ocular diseases such as uveal melanoma (UM). Poor prognosis of UM is generally associated with high potential of metastatic liver dissemination. A strong driver of metastatic dissemination is the activation of the epithelial-mesenchymal transition (EMT) regulating transcription factor ZEB1, and high expression of ZEB1 is associated with aggressiveness of UM. While ZEB1 expression can be also associated with immune tolerance, the underlying drivers of ZEB1 activation remain unclear. METHODS: Transcriptomic, in vitro, ex vivo, and in vivo analyses were used to investigate the impact on clinical prognosis of immune infiltration in the ocular tumor microenvironment. A metastatic liver dissemination model of was developed to address the role of natural killer (NK) cells in driving the migration of UM. RESULTS: In a pan-cancer TCGA analysis, natural killer (NK) cells were associated with worse overall survival in uveal melanoma and more abundant in high-risk monosomy 3 tumors. Furthermore, uveal melanoma expressed high levels of the tumor necrosis factor superfamily member 4-1BB ligand, particularly in tumors with monosomy 3 and BAP1 mutations. Tumors expressing 4-1BB ligand induced CD73 expression on NK cells accompanied with the ability to promote tumor dissemination. Through ligation of 4-1BB, NK cells induced the expression of the ZEB1 transcription factor, leading to the formation of liver metastasis of uveal melanoma. CONCLUSIONS: Taken together, the present study demonstrates a role of NK cells in the aggravation of uveal melanoma towards metastatic disease.


Subject(s)
4-1BB Ligand , Melanoma , Humans , Melanoma/genetics , Epithelial-Mesenchymal Transition , Killer Cells, Natural , Monosomy , Tumor Microenvironment
2.
Cancers (Basel) ; 13(5)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803245

ABSTRACT

Checkpoint inhibitors are slowly being introduced in the care of specific sarcoma subtypes such as undifferentiated pleomorphic sarcoma, alveolar soft part sarcoma, and angiosarcoma even though formal indication is lacking. Proper biomarkers to unravel potential immune reactivity in the tumor microenvironment are therefore expected to be highly warranted. In this study, intratumoral spatial cross presentation was investigated as a novel concept where immune cell composition in the tumor microenvironment was suggested to act as a proxy for immune surveillance. Double immunohistochemistry revealed a prognostic role of direct spatial interactions between CD11c+ antigen-presenting cells (APCs) and CD8+ cells in contrast to each marker alone in a soft tissue sarcoma (STS) cohort of 177 patients from the Karolinska University Hospital (MFS p = 0.048, OS p = 0.025). The survival benefit was verified in multivariable analysis (MFS p = 0.012, OS p = 0.004). Transcriptomics performed in the TCGA sarcoma cohort confirmed the prognostic value of combining CD11c with CD8 (259 patients, p = 0.005), irrespective of FOXP3 levels and in a CD274 (PD-LI)-rich tumor microenvironment. Altogether, this study presents a histopathological approach to link immune surveillance and patient survival in STS. Notably, spatial cross presentation as a prognostic marker is distinct from therapy response-predictive biomarkers such as immune checkpoint molecules of the PD-L1/PD1 pathway.

3.
Eur Neuropsychopharmacol ; 29(11): 1282-1287, 2019 11.
Article in English | MEDLINE | ID: mdl-31537475

ABSTRACT

Recently, a single injection of the nitric oxide donor sodium nitroprusside (SNP) was found to induce a rapid and sustained antipsychotic effect in treatment-resistant schizophrenia (TRS). Moreover, a single i.p. injection of SNP in rats was found to generate both rapid and persisting changes in brain synaptic plasticity, including enhanced excitatory postsynaptic current responses and spine morphology in layer V pyramidal cells in the medial prefrontal cortex (mPFC) brain slices. Here we used the conditioned avoidance response (CAR) test in rats to investigate the antipsychotic-like efficacy of SNP in combination with low-dose risperidone. In addition, we performed microdialysis experiments in freely moving rats to measure neurotransmitter efflux in the mPFC and the nucleus accumbens (NAc). Risperidone caused only 20% suppression of CAR, which is far below the degree of CAR suppression required to predict a significant clinical antipsychotic effect. Addition of a low dose of SNP to risperidone dramatically enhanced the antipsychotic-like effect to a clinically relevant level. SNP significantly enhanced the risperidone-induced dopamine output in the mPFC but not in the NAc. The increased prefrontal dopamine release induced by the drug combination may also improve cognition as indicated by previous preclinical and clinical studies and, furthermore, via enhanced synaptic spine function and morphology in mPFC generate a both rapid and prolonged antipsychotic and pro-cognitive effect. Our results delineate SNP as a promising new treatment option for schizophrenia, including TRS, when added to currently available antipsychotic medication in order to improve efficacy at maintained or even reduced dosage.


Subject(s)
Avoidance Learning/drug effects , Nitroprusside/pharmacology , Risperidone/pharmacology , Animals , Dopamine/metabolism , Drug Synergism , Male , Microdialysis , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Rats
4.
Eur Neuropsychopharmacol ; 26(9): 1401-1411, 2016 09.
Article in English | MEDLINE | ID: mdl-27474687

ABSTRACT

Nicotine has been found to improve cognition and reduce negative symptoms in schizophrenia and a genetic and pathophysiological link between the α7 nicotinic acetylcholine receptors (nAChRs) and schizophrenia has been demonstrated. Therefore, there has been a large interest in developing drugs affecting the α7 nAChRs for schizophrenia. In the present study we investigated, in rats, the effects of a selective α7 agonist (PNU282987) and a α7 positive allosteric modulator (PAM; NS1738) alone and in combination with the atypical antipsychotic drug risperidone for their utility as adjunct treatment in schizophrenia. Moreover we also investigated their utility as adjunct treatment in depression in combination with the SSRI citalopram. We found that NS1738 and to some extent also PNU282987, potentiated a subeffective dose of risperidone in the conditioned avoidance response test. Both drugs also potentiated the effect of a sub-effective concentration of risperidone on NMDA-induced currents in pyramidal cells of the medial prefrontal cortex. Moreover, NS1738 and PNU282987 enhanced recognition memory in the novel object recognition test, when given separately. Both drugs also potentiated accumbal but not prefrontal risperidone-induced dopamine release. Finally, PNU282987 reduced immobility in the forced swim test, indicating an antidepressant-like effect. Taken together, our data support the utility of drugs targeting the α7 nAChRs, perhaps especially α7 PAMs, to potentiate the effect of atypical antipsychotic drugs. Moreover, our data suggest that α7 agonists and PAMs can be used to ameliorate cognitive symptoms in schizophrenia and depression.


Subject(s)
Antidepressive Agents/pharmacology , Antipsychotic Agents/pharmacology , Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Phenylurea Compounds/pharmacology , Schizophrenia/drug therapy , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Citalopram/pharmacology , Depression/drug therapy , Depression/metabolism , Disease Models, Animal , Dopamine/metabolism , Excitatory Amino Acid Agonists/pharmacology , Male , N-Methylaspartate/metabolism , N-Methylaspartate/pharmacology , Nicotinic Agonists/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Risperidone/pharmacology , Schizophrenia/metabolism , Schizophrenic Psychology , Selective Serotonin Reuptake Inhibitors/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism
5.
Int J Neuropsychopharmacol ; 18(3)2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25522408

ABSTRACT

BACKGROUND: Substantial clinical data support the addition of low doses of atypical antipsychotic drugs to selective serotonin reuptake inhibitors (SSRIs) to rapidly enhance the antidepressant effect in treatment-resistant depression. Preclinical studies suggest that this effect is at least partly explained by an increased catecholamine outflow in the medial prefrontal cortex (mPFC). METHODS: In the present study we used in vivo microdialysis in freely moving rats and in vitro intracellular recordings of pyramidal cells of the rat mPFC to investigate the effects of adding the novel atypical antipsychotic drug asenapine to the SSRI escitalopram with regards to monoamine outflow in the mPFC and dopamine outflow in nucleus accumbens as well as glutamatergic transmission in the mPFC. RESULTS: The present study shows that addition of low doses (0.05 and 0.1 mg/kg) of asenapine to escitalopram (5 mg/kg) markedly enhances dopamine, noradrenaline, and serotonin release in the rat mPFC as well as dopamine release in the nucleus accumbens. Moreover, this drug combination facilitated both N-methyl-d-Aspartate (NMDA)- and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-induced currents as well as electrically evoked excitatory postsynaptic potentials in pyramidal cells of the rat mPFC. CONCLUSIONS: Our results support the notion that the augmentation of SSRIs by atypical antipsychotic drugs in treatment-resistant depression may, at least in part, be related to enhanced catecholamine output in the prefrontal cortex and that asenapine may be clinically used to achieve this end. In particular, the subsequent activation of the D1 receptor may be of importance for the augmented antidepressant effect, as this mechanism facilitated both NMDA and AMPA receptor-mediated transmission in the mPFC. Our novel observation that the drug combination, like ketamine, facilitates glutamatergic transmission in the mPFC may contribute to explain the rapid and potent antidepressant effect obtained when atypical antipsychotic drugs are added to SSRIs.


Subject(s)
Antipsychotic Agents/pharmacology , Biogenic Monoamines/metabolism , Citalopram/pharmacology , Glutamic Acid/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Prefrontal Cortex/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Synaptic Transmission/drug effects , Animals , Benzazepines/pharmacology , Bicuculline/pharmacology , Dibenzocycloheptenes , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , GABA-A Receptor Antagonists/pharmacology , Male , Neurons/drug effects , Prefrontal Cortex/cytology , Rats , Rats, Wistar , Time Factors
6.
Eur Neuropsychopharmacol ; 23(7): 709-20, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22732518

ABSTRACT

Quetiapine alleviates both positive and negative symptoms as well as certain cognitive impairments in schizophrenia despite a low D2 receptor occupancy and may also be used as monotherapy in bipolar and major depressive disorder. The mechanisms underlying the broad clinical utility of quetiapine remain to be clarified, but may be related to the potent inhibition of the norepinephrine transporter (NET) by norquetiapine, the major metabolite of quetiapine in humans. Since norquetiapine is not formed in rodents we here investigated in rats whether NET-inhibition may, in principle, contribute to the clinical effectiveness of quetiapine and allow for its low D2 receptor occupancy, by combining quetiapine with the selective NET-inhibitor reboxetine. Antipsychotic-like activity was assessed using the conditioned avoidance response (CAR) test, dopamine output in the medial prefrontal cortex (mPFC) and the nucleus accumbens was measured using in vivo microdialysis, and NMDA receptor-mediated transmission was measured using intracellular electrophysiological recordings in pyramidal cells of the mPFC in vitro. Adjunct reboxetine potentiated the suppression of CAR by quetiapine. Moreover, concomitant administration of quetiapine and reboxetine resulted in a synergistic increase in cortical, but not accumbal, dopamine output. The combination of low, clinically relevant concentrations of quetiapine (60 nM) and reboxetine (20 nM) markedly facilitated cortical NMDA receptor-mediated transmission in contrast to either drug alone, an effect that could be inhibited by the D1 receptor antagonist SCH23390. We conclude that concomitant NET-inhibition by norquetiapine may contribute to the overall antipsychotic effectiveness of quetiapine in spite of its relatively low level of D2 occupancy.


Subject(s)
Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Dibenzothiazepines/pharmacology , Drug Synergism , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , Benzazepines/pharmacology , Dibenzothiazepines/antagonists & inhibitors , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Male , Membrane Potentials/drug effects , Morpholines/antagonists & inhibitors , Morpholines/pharmacology , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/pharmacology , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Quetiapine Fumarate , Raclopride/pharmacology , Rats , Reboxetine
7.
Psychopharmacology (Berl) ; 221(1): 115-31, 2012 May.
Article in English | MEDLINE | ID: mdl-22068461

ABSTRACT

RATIONALE: The α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor positive allosteric modulators (AMPA-PAMs), Org 24448 and Org 26576, and the glycine transporter-1 (GlyT-1) inhibitor Org 25935 are developed for treatment of schizophrenia. OBJECTIVES: Here we examined experimentally the ability of co-administration of these AMPA-PAMs or the GlyT-1 inhibitor to augment the antipsychotic activity and effect on cortical N-methyl-D: -aspartate (NMDA) receptor-mediated transmission of risperidone, olanzapine, or haloperidol. METHODS: We examined antipsychotic efficacy using the conditioned avoidance response (CAR) test, extrapyramidal side effect liability using a catalepsy test, and cortical NMDA receptor-mediated glutamatergic transmission using intracellular electrophysiological recording technique in vitro. RESULTS: Both AMPA-PAMs enhanced the suppression of CAR induced by risperidone or olanzapine, and Org 24448 also enhanced the effect of haloperidol. In contrast, the GlyT-1 inhibitor did not cause any behaviorally significant effect in the CAR test. However, the GlyT-1 inhibitor, but not the AMPA-PAMs, produced a large facilitation of NMDA-induced currents. All three drugs potentiated the effect of risperidone but not haloperidol on these currents. The GlyT-1 inhibitor also facilitated the effect of olanzapine. All drugs potentiated the effect of risperidone on electrically stimulated excitatory postsynaptic potentials (EPSP) in cortical pyramidal cells, whereas only the GlyT inhibitor facilitated the effect of olanzapine. CONCLUSIONS: Our results suggest that the AMPA-PAMs, when compared to the GlyT-1 inhibitor, show differential effects in terms of augmentation of antipsychotic efficacy, particularly when combined with risperidone or olanzapine. Both AMPA-PAMs and the GlyT-1 inhibitor may also improve negative symptoms and cognitive impairments in schizophrenia, in particular when combined with risperidone.


Subject(s)
Antipsychotic Agents/agonists , Avoidance Learning/physiology , Glutamic Acid/metabolism , Oxadiazoles/pharmacology , Piperidines/pharmacology , Prefrontal Cortex/physiology , Receptors, AMPA/agonists , Risperidone/pharmacology , Synaptic Transmission/physiology , Animals , Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Benzodiazepines/agonists , Benzodiazepines/pharmacology , Catalepsy/physiopathology , Drug Synergism , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Haloperidol/agonists , Haloperidol/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Olanzapine , Oxadiazoles/administration & dosage , Piperidines/administration & dosage , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Risperidone/agonists , Synaptic Transmission/drug effects , Tetrahydronaphthalenes/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/administration & dosage , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/analogs & derivatives , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
8.
Synapse ; 66(4): 277-90, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22121030

ABSTRACT

Antidepressant drugs are frequently used to treat affective symptoms in schizophrenia. We have recently shown that escitalopram, but not citalopram or R-citalopram, increases firing rate and burst firing of midbrain dopamine neurons, potentiates cortical N-methyl-D-aspartate (NMDA) receptor-mediated transmission and enhances cognition, effects that might influence the outcome of concomitant antipsychotic medication. Here, we studied, in rats, the behavioral and neurobiological effects of adding escitalopram, citalopram, or R-citalopram to the second-generation antipsychotic drug risperidone. We examined antipsychotic efficacy using the conditioned avoidance response (CAR) test, extrapyramidal side effect (EPS) liability using a catalepsy test, dopamine outflow in the medial prefrontal cortex (mPFC) and nucleus accumbens using in vivo microdialysis in freely moving animals, and NMDA receptor-mediated transmission in the mPFC using intracellular electrophysiological recording in vitro. Only escitalopram (5 mg/kg), but not citalopram (10 mg/kg), or R-citalopram (10 mg/kg), dramatically enhanced the antipsychotic-like effect of a low dose of risperidone (0.25 mg/kg), without increasing catalepsy. Given alone, escitalopram, but not citalopram or R-citalopram, markedly enhanced both cortical dopamine output and NMDA receptor-mediated transmission. Addition of escitalopram and to some extent R-citalopram, but not citalopram, significantly enhanced both cortical dopamine output and cortical NMDA receptor-mediated transmission induced by a suboptimal dose/concentration of risperidone. These results suggest that adjunct treatment with escitalopram, but not citalopram, may enhance the effect of a subtherapeutic dose of risperidone on positive, negative, cognitive, and depressive symptoms in schizophrenia, yet without increased EPS liability.


Subject(s)
Antipsychotic Agents/administration & dosage , Brain/drug effects , Citalopram/administration & dosage , Risperidone/administration & dosage , Selective Serotonin Reuptake Inhibitors/administration & dosage , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Brain/metabolism , Chromatography, High Pressure Liquid , Dopamine/metabolism , Drug Synergism , Drug Therapy, Combination , Electrophysiological Phenomena/drug effects , Male , Microdialysis , Patch-Clamp Techniques , Rats , Rats, Wistar
9.
Neuropsychopharmacology ; 35(9): 1952-61, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20463659

ABSTRACT

Preclinical data have shown that addition of the selective norepinephrine transporter (NET) inhibitor reboxetine increases the antipsychotic-like effect of the D(2/3) antagonist raclopride and, in parallel, enhances cortical dopamine output. Subsequent clinical results suggested that adding reboxetine to stable treatments with various antipsychotic drugs (APDs) may improve positive, negative and depressive symptoms in schizophrenia. In this study, we investigated in rats the effects of adding reboxetine to the second-generation APD olanzapine on: (i) antipsychotic efficacy, using the conditioned avoidance response (CAR) test, (ii) extrapyramidal side effect (EPS) liability, using a catalepsy test, (iii) dopamine efflux in the medial prefrontal cortex and the nucleus accumbens, using in vivo microdialysis in freely moving animals and (iv) cortical N-methyl-D-aspartate (NMDA) receptor-mediated transmission, using intracellular electrophysiological recording in vitro. Reboxetine (6 mg/kg) enhanced the suppression of CAR induced by a suboptimal dose (1.25 mg/kg), but not an optimal (2.5 mg/kg) dose of olanzapine without any concomitant catalepsy. Addition of reboxetine to the low dose of olanzapine also markedly increased cortical dopamine outflow and facilitated prefrontal NMDA receptor-mediated transmission. Our data suggest that adjunctive treatment with a NET inhibitor may enhance the therapeutic effect of low-dose olanzapine in schizophrenia without increasing EPS liability and add an antidepressant action, thus in principle allowing for a dose reduction of olanzapine with a concomitant reduction of dose-related side effects, such as EPS and weight gain.


Subject(s)
Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Benzodiazepines/pharmacology , Cerebral Cortex/drug effects , Dopamine/metabolism , Morpholines/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Behavior, Animal/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Excitatory Amino Acid Agonists/pharmacology , Freezing Reaction, Cataleptic/drug effects , In Vitro Techniques , Male , Membrane Potentials/drug effects , Microdialysis/methods , N-Methylaspartate/pharmacology , Olanzapine , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reboxetine
10.
Vaccine ; 28(10): 2161-2168, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20060511

ABSTRACT

The aim of the present study was to synthesise and screen a set of novel nicotine hapten immunogens used for the treatment of nicotine dependence. In the screening process we studied the amount of antibodies generated and their selectivity, using ELISA techniques, and their effects on nicotine-induced dopamine release in the NAC(shell) of the rat, assessed by in vivo voltammetry. We conclude that even small changes such as the linker attachment on the nicotine molecule as well as the structure of the linker may greatly influence the selectivity of the antibodies and the central neurobiological effects of nicotine that are considered critical for its dependence producing properties.


Subject(s)
Drug Evaluation, Preclinical/methods , Haptens/immunology , Nicotine/immunology , Substance-Related Disorders/therapy , Vaccines/therapeutic use , Animals , Antibodies/blood , Dopamine/metabolism , Enzyme-Linked Immunosorbent Assay , Haptens/chemistry , Male , Molecular Structure , Nicotine/chemistry , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...