Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Anal Chem ; 94(4): 1932-1940, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34965097

RESUMO

Glioblastoma multiforme (GBM) is the most aggressive brain tumor, characterized by short median survival and an almost 100% tumor-related mortality. The standard of care treatment for newly diagnosed GBM includes surgical resection followed by concomitant radiochemotherapy. The prevention of disease progression fails due to the poor therapeutic effect caused by the great molecular heterogeneity of this tumor. Previously, we exploited synchrotron radiation-based soft X-ray tomography and hard X-ray fluorescence for elemental microimaging of the shock-frozen GBM cells. The present study focuses instead on the biochemical profiling of live GBM cells and provides new insight into tumor heterogenicity. We studied bio-macromolecular changes by exploring the live-cell synchrotron-based Fourier transform infrared (SR-FTIR) microspectroscopy in a set of three GBM cell lines, including the patient-derived glioblastoma cell line, before and after riluzole treatment, a medicament with potential anticancer properties. SR-FTIR microspectroscopy shows that GBM live cells of different origins recruit different organic compounds. The riluzole treatment of all GBM cell lines mainly affected carbohydrate metabolism and the DNA structure. Lipid structures and protein secondary conformation are affected as well by the riluzole treatment: cellular proteins assumed cross ß-sheet conformation while parallel ß-sheet conformation was less represented for all GBM cells. Moreover, we hope that a new live-cell approach for GBM simultaneous treatment and examination can be devised to target cancer cells more specifically, i.e., future therapies can develop more specific treatments according to the specific bio-macromolecular signature of each tumor type.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Encéfalo/metabolismo , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Glioblastoma/diagnóstico por imagem , Glioblastoma/tratamento farmacológico , Humanos , Riluzol/uso terapêutico , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Síncrotrons
2.
Acta Neurochir (Wien) ; 161(5): 1037-1045, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30877471

RESUMO

BACKGROUND: Brain metastases (BMs) are the most frequent malignancy of the central nervous system. Previous research suggested that some metastases show infiltrative behavior rather than sharp demarcation. We hypothesized that three magnetic resonance (MR) imaging parameters-(a) tumor size, (b) extent of peritumoral edema, and (c) presence of multiple BMs-are predictors of cellular invasion beyond the surgically identifiable tumor margins. METHODS: We performed a post hoc analysis on prospectively collected data of patients with BMs. Biopsies beyond the resection margin and immunohistochemistry were performed to assess infiltration status. The three MR imaging parameters were dichotomized into diameters ≤ 30 mm ("small") and > 30 mm ("large"), amount of peritumoral edema "extended" and "limited," and "multiple BMs" and "single BMs," respectively. The association between infiltration status and imaging parameters was calculated using chi-square test. RESULTS: Biopsy beyond the resection margin was performed in 77 patients; 49 (63.6%) had supramarginal infiltration and 28 patients (36.4%) showed no infiltration. Histological evidence of tumor infiltration was found in 25/41 patients with smaller lesions (61%) and in 24/36 with larger lesions (66.7%, p = 0.64), in 28/44 patients with limited (63.6%) and in 21/33 patients with extended edema (63.6%, p = 1.0), in 28/45 patients (62.2%) with single BM and in 21/32 patients (65.6%) with multiple BMs (p = 0.81). CONCLUSIONS: Based on the post hoc analysis of our prospective trial data, we could not confirm the hypothesis that infiltration of brain parenchyma beyond the glial pseudocapsule is associated with the MR imaging parameters tumor size, extent of edema, or multiplicity of metastases.


Assuntos
Edema Encefálico/diagnóstico por imagem , Neoplasias Encefálicas/diagnóstico por imagem , Adulto , Idoso , Edema Encefálico/epidemiologia , Edema Encefálico/patologia , Neoplasias Encefálicas/epidemiologia , Neoplasias Encefálicas/patologia , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica
3.
Anticancer Res ; 44(5): 1829-1835, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38677733

RESUMO

BACKGROUND/AIM: Glioblastoma multiforme (GBM)-induced oedema is a major cause of morbidity and mortality among patients with GBM. Dexamethasone (Dex) is the most common corticosteroid used pre-operatively to control cerebral oedema in patients with GBM. Dex is associated with many side effects, and shorter overall survival and progression-free survival of patients with GBM. These negative effects of Dex highlight the need for combinational therapy. Riluzole (Ril), a drug used to treat amyotrophic lateral sclerosis (ALS), is thought to have potential as a treatment for various cancers, with clinical trials underway. Here, we investigated whether Ril could reverse some of the undesirable effects of Dex. MATERIALS AND METHODS: The effect of Dex, Ril, and Ril-Dex treatment on cell migration was monitored using the xCELLigence system. Cell viability assays were performed using 3-(4, 5-dimethylthiazol)-2, 5-diphenyltetrazolium bromide (MTT). The expression of genes involved in migration, glucose metabolism, and stemness was examined using real-time polymerase chain reaction (PCR). RESULTS: Pre-treating GBM cells with Ril reduced Dex-induced cell migration and altered Dex-induced effects on cell invasion, stem cell, and glucose metabolism markers. Furthermore, Ril remained effective in killing GBM cells in combination with Dex. CONCLUSION: Ril, which acts as an anti-tumorigenic drug, mediates some of the negative effects of Dex; therefore, it could be a potential drug to manage the side effects of Dex therapy in GBM.


Assuntos
Movimento Celular , Dexametasona , Glioblastoma , Riluzol , Riluzol/farmacologia , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/metabolismo , Dexametasona/farmacologia , Movimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Sobrevivência Celular/efeitos dos fármacos
4.
Hum Mol Genet ; 20(20): 4076-81, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21791550

RESUMO

Common sequence variants have recently joined rare structural polymorphisms as genetic factors with strong evidence for association with schizophrenia. Here we extend our previous genome-wide association study and meta-analysis (totalling 7 946 cases and 19 036 controls) by examining an expanded set of variants using an enlarged follow-up sample (up to 10 260 cases and 23 500 controls). In addition to previously reported alleles in the major histocompatibility complex region, near neurogranin (NRGN) and in an intron of transcription factor 4 (TCF4), we find two novel variants showing genome-wide significant association: rs2312147[C], upstream of vaccinia-related kinase 2 (VRK2) [odds ratio (OR) = 1.09, P = 1.9 × 10(-9)] and rs4309482[A], between coiled-coiled domain containing 68 (CCDC68) and TCF4, about 400 kb from the previously described risk allele, but not accounted for by its association (OR = 1.09, P = 7.8 × 10(-9)).


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Polimorfismo de Nucleotídeo Único , Proteínas Serina-Treonina Quinases/genética , Esquizofrenia/genética , Fatores de Transcrição/genética , Alelos , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Risco , Fator de Transcrição 4
5.
Mol Med ; 18: 628-35, 2012 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-22396019

RESUMO

Erythropoietin (EPO) reduces symptoms of experimental autoimmune encephalomyelitis in rodents and shows neuroregenerative effects in chronic progressive multiple sclerosis. The mechanisms of action of EPO in these conditions with shared immunological etiology are still unclear. Therefore, we used a model of toxic demyelination allowing exclusion of T cell-mediated inflammation. In a double-blind (for food/injections), placebo-controlled, longitudinal four-arm design, 8-wk-old C57BL/6 mice (n = 26/group) were assigned to cuprizone-containing (0.2%) or regular food (ground chow) for 6 wks. After 3 wks, mice were injected every other day with placebo or EPO (5,000 IU/kg intraperitoneally) until the end of cuprizone feeding. Half of the mice were exposed to behavioral testing, magnetic resonance imaging (MRI) and histology immediately after treatment cessation, whereas the other half were allowed a 3-wk treatment-free recovery. Immediately after termination of cuprizone feeding, all toxin-exposed mice were compromised regarding vestibulomotor function/coordination, with EPO-treated animals performing better than placebo. Likewise, ventricular enlargement after cuprizone, as documented by MRI, was less pronounced upon EPO. After a 3-wk recovery, remarkable spontaneous improvement was observed in all mice with no measurable further benefit in the EPO group ("ceiling effect"). Histological analysis of the corpus callosum revealed attenuation by EPO of the cuprizone-induced increase in microglial numbers and amyloid precursor protein accumulations as a readout of inflammation and axonal degeneration. To conclude, EPO ameliorates neurological symptoms in the cuprizone model of demyelination, possibly by reduction of inflammation-associated axonal degeneration in white matter tracts. These findings underscore the value of future therapeutic strategies for multiple sclerosis based on EPO or EPO variants.


Assuntos
Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/patologia , Eritropoetina/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Axônios/patologia , Ventrículos Cerebrais/patologia , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/patologia , Cuprizona , Doenças Desmielinizantes/induzido quimicamente , Eritropoetina/administração & dosagem , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Fármacos Neuroprotetores/administração & dosagem , Equilíbrio Postural , Desempenho Psicomotor
6.
Mol Med ; 18: 1029-40, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22669473

RESUMO

Erythropoietin (EPO) improves cognitive performance in clinical studies and rodent experiments. We hypothesized that an intrinsic role of EPO for cognition exists, with particular relevance in situations of cognitive decline, which is reflected by associations of EPO and EPO receptor (EPOR) genotypes with cognitive functions. To prove this hypothesis, schizophrenic patients (N > 1000) were genotyped for 5' upstream-located gene variants, EPO SNP rs1617640 (T/G) and EPORSTR(GA)(n). Associations of these variants were obtained for cognitive processing speed, fine motor skills and short-term memory readouts, with one particular combination of genotypes superior to all others (p < 0.0001). In an independent healthy control sample (N > 800), these associations were confirmed. A matching preclinical study with mice demonstrated cognitive processing speed and memory enhanced upon transgenic expression of constitutively active EPOR in pyramidal neurons of cortex and hippocampus. We thus predicted that the human genotypes associated with better cognition would reflect gain-of-function effects. Indeed, reporter gene assays and quantitative transcriptional analysis of peripheral blood mononuclear cells showed genotype-dependent EPO/EPOR expression differences. Together, these findings reveal a role of endogenous EPO/EPOR for cognition, at least in schizophrenic patients.


Assuntos
Cognição , Eritropoetina/genética , Predisposição Genética para Doença , Polimorfismo Genético , Receptores da Eritropoetina/genética , Esquizofrenia/genética , Esquizofrenia/fisiopatologia , Adolescente , Adulto , Idoso , Animais , Estudos de Casos e Controles , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Demografia , Feminino , Estudos de Associação Genética , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Masculino , Memória , Camundongos , Pessoa de Meia-Idade , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Células Piramidais/metabolismo , Células Piramidais/patologia , Adulto Jovem
7.
BMC Biol ; 9: 27, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21527022

RESUMO

BACKGROUND: Erythropoietin (EPO) and its receptor (EPOR) are expressed in the developing brain and their transcription is upregulated in adult neurons and glia upon injury or neurodegeneration. We have shown neuroprotective effects and improved cognition in patients with neuropsychiatric diseases treated with EPO. However, the critical EPO targets in brain are unknown, and separation of direct and indirect effects has remained difficult, given the role of EPO in hematopoiesis and brain oxygen supply. RESULTS: Here we demonstrate that mice with transgenic expression of a constitutively active EPOR isoform (cEPOR) in pyramidal neurons of cortex and hippocampus exhibit enhancement of spatial learning, cognitive flexibility, social memory, and attentional capacities, accompanied by increased impulsivity. Superior cognitive performance is associated with augmented long-term potentiation of cEPOR expressing neurons in hippocampal slices. CONCLUSIONS: Active EPOR stimulates neuronal plasticity independent of any hematopoietic effects and in addition to its neuroprotective actions. This property of EPOR signaling should be exploited for defining novel strategies to therapeutically enhance cognitive performance in disease conditions.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Cognição/efeitos dos fármacos , Eritropoetina/farmacologia , Hipocampo/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Receptores da Eritropoetina/metabolismo , Animais , Animais Geneticamente Modificados , Atenção , Córtex Cerebral/fisiologia , Eritropoetina/metabolismo , Hipocampo/fisiologia , Humanos , Comportamento Impulsivo , Aprendizagem , Masculino , Memória , Camundongos , Células Piramidais/fisiologia , Receptores da Eritropoetina/genética , Proteínas Recombinantes , Comportamento Social
8.
Transl Stroke Res ; 13(4): 616-624, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35061211

RESUMO

Cerebral vasospasm is a highly investigated phenomenon in neurovascular research. Experimental vasospasm models are irreplaceable for the evaluation of new antivasospastic drugs. In this study, we assessed the reliability of in vivo vasospasm induction by ultrasound application in the chicken chorioallantoic membrane (CAM) model. After incubation of fertilized chicken eggs for four days, a fenestration was performed to enable examination of the CAM vessels. On the thirteenth day, continuous-wave ultrasound (3 MHz, 1 W/cm2) was applied on the CAM vessels for 60 s. The ultrasound effect on the vessels was recorded by life imaging (5-MP HD-microscope camera, Leica®). The induced vessel diameter changes were evaluated in a defined time interval of 20 min using a Fiji macro. The vessel diameter before and after sonication was measured and the relative diameter reduction was determined. A first reduction of vessel diameter was observed after three minutes with an average vessel-diameter decrease to 77%. The maximum reduction in vessel diameter was reached eight minutes after sonication with an average vessel diameter decrease to 57% (mean relative diameter reduction of 43%, range 44-61%), ANOVA, p = 0.0002. The vasospasm persisted for all 20 recorded minutes post induction. Vasospasm can be reliably induced by short application of 3 MHz-ultrasound to the CAM vessels. This might be a suitable in vivo model for the evaluation of drug effects on vasospasm in an experimental setting as intermediary in the transition process from in vitro to in vivo assessment using animal models.


Assuntos
Membrana Corioalantoide , Vasoespasmo Intracraniano , Animais , Galinhas , Membrana Corioalantoide/irrigação sanguínea , Reprodutibilidade dos Testes , Ultrassonografia
9.
Transl Stroke Res ; 13(5): 792-800, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34988870

RESUMO

Nimodipine prevents cerebral vasospasm and improves functional outcome after aneurysmal subarachnoid hemorrhage (aSAH). The beneficial effect is limited by low oral bioavailability of nimodipine, which resulted in an increasing use of nanocarriers with sustained intrathecal drug release in order to overcome this limitation. However, this approach facilitates only a continuous and not an on-demand nimodipine release during the peak time of vasospasm development. In this study, we aimed to assess the concept of controlled drug release from nimodipine-loaded copolymers by ultrasound application in the chicken chorioallantoic membrane (CAM) model. Nimodipine-loaded copolymers were produced with the direct dissolution method. Vasospasm of the CAM vessels was induced by means of ultrasound (Physiomed, continuous wave, 3 MHz, 1.0 W/cm2). The ultrasound-mediated nimodipine release (Physiomed, continuous wave, 1 MHz, 1.7 W/cm2) and its effect on the CAM vessels were evaluated. Measurements of vessel diameter before and after ultrasound-induced nimodipine release were performed using ImageJ. The CAM model could be successfully carried out in all 25 eggs. After vasospasm induction and before drug release, the mean vessel diameter was at 57% (range 44-61%) compared to the baseline diameter (set at 100%). After ultrasound-induced drug release, the mean vessel diameter of spastic vessels increased again to 89% (range 83-91%) of their baseline diameter, which was significant (p = 0.0002). We were able to provide a proof of concept for in vivo vasospasm induction by ultrasound application in the CAM model and subsequent resolution by ultrasound-mediated nimodipine release from nanocarriers. This concept merits further evaluation in a rat SAH model.


Assuntos
Hemorragia Subaracnóidea , Vasoespasmo Intracraniano , Animais , Micelas , Nimodipina , Ratos , Ultrassonografia , Vasodilatadores , Vasoespasmo Intracraniano/diagnóstico por imagem , Vasoespasmo Intracraniano/tratamento farmacológico , Vasoespasmo Intracraniano/etiologia
10.
Mol Med ; 17(11-12): 1306-10, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21912808

RESUMO

The German Multicenter EPO Stroke Trial, which investigated safety and efficacy of erythropoietin (EPO) treatment in ischemic stroke, was formally declared a negative study. Exploratory subgroup analysis, however, revealed that patients not receiving thrombolysis most likely benefited from EPO during clinical recovery, a result demonstrated in the findings of the Göttingen EPO Stroke Study. The present work investigated whether the positive signal on clinical outcome in this patient subgroup was mirrored by respective poststroke biomarker profiles. All patients of the German Multicenter EPO Stroke Trial nonqualifying for thrombolysis were included if they (a) were treated per protocol and (b) had at least two of the five follow-up blood samples for circulating damage markers drawn (n = 163). The glial markers S100B and glial fibrillary acid protein (GFAP) and the neuronal marker ubiquitin C-terminal hydrolase (UCH-L1) were measured by enzyme-linked immunosorbent assay in serum on d 1, 2, 3, 4 and 7 poststroke. All biomarkers increased poststroke. Overall, EPO-treated patients had significantly lower concentrations (area under the curve) over 7 d of observation, as reflected by the composite score of all three markers (Cronbach α = 0.811) and by UCH-L1. S100B and GFAP showed a similar tendency. To conclude, serum biomarker profiles, as an outcome measure of brain damage, corroborate an advantageous effect of EPO in ischemic stroke. In particular, reduction in the neuronal damage marker UCH-L1 may reflect neuroprotection by EPO.


Assuntos
Isquemia Encefálica/sangue , Isquemia Encefálica/tratamento farmacológico , Eritropoetina/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/tratamento farmacológico , Idoso , Área Sob a Curva , Biomarcadores/sangue , Isquemia Encefálica/complicações , Epoetina alfa , Eritropoetina/farmacologia , Feminino , Humanos , Masculino , Fármacos Neuroprotetores/farmacologia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Acidente Vascular Cerebral/complicações , Resultado do Tratamento
11.
Glia ; 57(7): 693-702, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18985736

RESUMO

A small experimental cryolesion to the right parietal cortex of juvenile mice causes late-onset global brain atrophy with memory impairments, reminiscent of cognitive decline, and progressive brain matter loss in schizophrenia. However, the cellular events underlying this global neurodegeneration are not understood. Here we show, based on comprehensive stereological analysis, that early unilateral lesion causes immediate and lasting bilateral increase in the number of microglia in cingulate cortex and hippocampus, consistent with a chronic low-grade inflammatory process. Whereas the total number of neurons and astrocytes in these brain regions remain unaltered, pointing to a non- gliotic neurodegeneration (as seen in schizophrenia), the subgroup of parvalbumin-positive inhibitory GABAergic interneurons is increased bilaterally in the hippocampus, as is the expression of the GABA-synthesizing enzyme GAD67. Moreover, unilateral parietal lesion causes a decrease in the expression of synapsin1, suggesting impairment of presynaptic functions/neuroplasticity. Reduced expression of the myelin protein cyclic nucleotide phosphodiesterase, reflecting a reduction of oligodendrocytes, may further contribute to the observed brain atrophy. Remarkably, early intervention with recombinant human erythropoietin (EPO), a hematopoietic growth factor with multifaceted neuroprotective properties (intraperitoneal injection of 5000 IU/kg body weight every other day for 3 weeks), prevented all these neurodegenerative changes. To conclude, unilateral parietal lesion of juvenile mice induces a non- gliotic neurodegenerative process, susceptible to early EPO treatment. Although the detailed mechanisms remain to be defined, these profound EPO effects open new ways for prophylaxis and therapy of neuropsychiatric diseases, e.g. schizophrenia.


Assuntos
Lesões Encefálicas/complicações , Encéfalo/fisiopatologia , Gliose/complicações , Gliose/fisiopatologia , Doenças Neurodegenerativas/complicações , Doenças Neurodegenerativas/fisiopatologia , Animais , Astrócitos/fisiologia , Atrofia , Encéfalo/patologia , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Lesões Encefálicas/terapia , Temperatura Baixa , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Modelos Animais de Doenças , Eritropoetina/uso terapêutico , Gliose/patologia , Gliose/terapia , Glutamato Descarboxilase/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/fisiologia , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/terapia , Neurônios/fisiologia , Parvalbuminas/metabolismo , Sinapsinas/metabolismo , Ácido gama-Aminobutírico/metabolismo
12.
BMC Biol ; 6: 37, 2008 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-18782446

RESUMO

BACKGROUND: Erythropoietin (EPO) improves cognition of human subjects in the clinical setting by as yet unknown mechanisms. We developed a mouse model of robust cognitive improvement by EPO to obtain the first clues of how EPO influences cognition, and how it may act on hippocampal neurons to modulate plasticity. RESULTS: We show here that a 3-week treatment of young mice with EPO enhances long-term potentiation (LTP), a cellular correlate of learning processes in the CA1 region of the hippocampus. This treatment concomitantly alters short-term synaptic plasticity and synaptic transmission, shifting the balance of excitatory and inhibitory activity. These effects are accompanied by an improvement of hippocampus dependent memory, persisting for 3 weeks after termination of EPO injections, and are independent of changes in hematocrit. Networks of EPO-treated primary hippocampal neurons develop lower overall spiking activity but enhanced bursting in discrete neuronal assemblies. At the level of developing single neurons, EPO treatment reduces the typical increase in excitatory synaptic transmission without changing the number of synaptic boutons, consistent with prolonged functional silencing of synapses. CONCLUSION: We conclude that EPO improves hippocampus dependent memory by modulating plasticity, synaptic connectivity and activity of memory-related neuronal networks. These mechanisms of action of EPO have to be further exploited for treating neuropsychiatric diseases.


Assuntos
Eritropoetina/farmacologia , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Memória/efeitos dos fármacos , Animais , Técnicas de Cultura de Células , Células Cultivadas , Eletrofisiologia , Hipocampo/fisiologia , Immunoblotting , Potenciação de Longa Duração/fisiologia , Masculino , Memória/fisiologia , Camundongos , Microscopia Confocal , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinapses/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/fisiologia
13.
Anticancer Res ; 39(1): 207-214, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30591460

RESUMO

BACKGROUND/AIM: Glioblastoma multiforme (GBM) is a malignant primary brain tumor with high rates of recurrence. This study aimed to investigate the effect of repurposed drug combinations on GBM. MATERIALS AND METHODS: Viability of U87 MG and 11ST patient-derived GMB cell lines, after valproic acid, tranylcypromine or riluzole alone, in different combinations, as well as combined with standard temozolomide chemotherapy was examined using the MTT assay. Proliferation, mRNA level of tissue factor pathway inhibitor 2 (TFPI2), and cell invasion were evaluated using anti-Ki-67 antibody staining, reverse transcriptase-polymerase chain reaction and xCELLigence system. RESULTS: The strongest effect on cell viability was achieved by the combination of riluzole with valproic acid (U87MG: 27.2%, 11ST: 25.99%). Tranylcypromine significantly enhanced the effect of temozolomide when used in combination, as did valproic acid. The normally high proliferation of GBM significantly declined under treatment with valproic acid with tranylcypromine (p=0.01). Finally, we observed reduction of invasion comparing single tranylcypromine to its combination with valproic acid or riluzole. CONCLUSION: These results support the idea that combinations of drugs could increase the treatment efficiency of GBM.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Glioblastoma/tratamento farmacológico , Glicoproteínas/genética , Recidiva Local de Neoplasia/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Riluzol/administração & dosagem , Temozolomida/administração & dosagem , Tranilcipromina/administração & dosagem , Ácido Valproico/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Mol Med ; 14(9-10): 546-52, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18552976

RESUMO

Molecular mechanisms underlying bipolar affective disorders are unknown. Difficulties arise from genetic and phenotypic heterogeneity of patients and the lack of animal models. Thus, we focused on only one patient (n = 1) with an extreme form of rapid cycling. Ribonucleic acid (RNA) from peripheral blood mononuclear cells (PBMC) was analyzed in a three-tiered approach under widely standardized conditions. Firstly, RNA was extracted from PBMC of eight blood samples, obtained on two consecutive days within one particular episode, including two different consecutive depressive and two different consecutive manic episodes, and submitted to (1) screening by microarray hybridizations, followed by (2) detailed bioinformatic analysis, and (3) confirmation of episode-specific regulation of genes by quantitative real-time polymerase chain reaction (qRT-PCR).Secondly, results were validated in additional blood samples obtained one to two years later. Among gene transcripts elevated in depressed episodes were prostaglandin D synthetase (PTGDS) and prostaglandin D2 11-ketoreductase (AKR1C3), both involved in hibernation. We hypothesized them to account for some of the rapid cycling symptoms. A subsequent treatment approach over 5 months applying the cyclooxygenase inhibitor celecoxib (2 x 200 mg daily) resulted in reduced severity rating of both depressed and manic episodes. This case suggests that rapid cycling is a systemic disease, resembling hibernation, with prostaglandins playing a mediator role.


Assuntos
3-Hidroxiesteroide Desidrogenases , Transtorno Bipolar/tratamento farmacológico , Inibidores de Ciclo-Oxigenase , Regulação da Expressão Gênica , Hidroxiprostaglandina Desidrogenases , Oxirredutases Intramoleculares , Leucócitos Mononucleares/metabolismo , Lipocalinas , Pirazóis , Sulfonamidas , 3-Hidroxiesteroide Desidrogenases/genética , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Transtorno Bipolar/diagnóstico , Transtorno Bipolar/genética , Transtorno Bipolar/metabolismo , Celecoxib , Biologia Computacional , Inibidores de Ciclo-Oxigenase/administração & dosagem , Inibidores de Ciclo-Oxigenase/uso terapêutico , Feminino , Perfilação da Expressão Gênica , Humanos , Hidroxiprostaglandina Desidrogenases/genética , Hidroxiprostaglandina Desidrogenases/metabolismo , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Lipocalinas/genética , Lipocalinas/metabolismo , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas/genética , Proteínas/metabolismo , Pirazóis/administração & dosagem , Pirazóis/uso terapêutico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfonamidas/administração & dosagem , Sulfonamidas/uso terapêutico , Resultado do Tratamento
15.
Brain ; 129(Pt 2): 480-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16339796

RESUMO

In humans, neurotrauma is suspected to cause brain atrophy and accelerate slowly progressive neurodegenerative disorders, such as Alzheimer's disease or schizophrenia. However, a direct link between brain injury and subsequent delayed global neurodegeneration has remained elusive. Here we show that juvenile (4-week-old) mice that are given a discrete unilateral lesion of the parietal cortex, develop to adulthood without obvious clinical symptoms. However, when monitored 3 and 9 months after lesioning, using high-resolution three-dimensional MRI and behavioural testing, the same mice display global neurodegenerative changes. Surprisingly, erythropoietin, a haematopoietic growth factor with potent neuroprotective activity, prevents behavioural abnormalities, cognitive dysfunction and brain atrophy when given for 2 weeks after acute brain injury. This demonstrates that a localized brain lesion is a primary cause of delayed global neurodegeneration that can be efficiently counteracted by neuroprotection.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/patologia , Encéfalo/patologia , Eritropoetina/uso terapêutico , Doenças Neurodegenerativas/prevenção & controle , Doença Aguda , Animais , Atrofia , Lesões Encefálicas/complicações , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Doenças Neurodegenerativas/diagnóstico , Doenças Neurodegenerativas/etiologia , Testes Neuropsicológicos , Fatores de Tempo
16.
Oncotarget ; 8(57): 96697-96709, 2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-29228563

RESUMO

A small subpopulation of tumor stem-like cells has the capacity to initiate tumors and mediate radio- and chemoresistance in diverse cancers hence also in glioblastoma (GBM). It has been reported that this capacity of tumor initiation in the brain is mainly dependent on the body's nutrient supply. This population of so-called brain tumor initiating or brain tumor stem-like cells (BTSCs) is able to extract nutrients like glucose with a higher affinity. Riluzole, a drug approved for treating amyotrophic lateral sclerosis (ALS), was reported to possess anticancer properties, affecting the glutamate metabolism. We report that riluzole treatment inhibits the growth of brain tumor stem-like cells enriched cultures isolated from two human glioblastomas. The effects of riluzole on these cells were associated with an inhibition of a poor prognostic indicator: glucose transporter 3 (GLUT3). A decrease in GLUT3 is associated with a decrease in the p-Akt/HIF1α pathway. Further, downregulation of the DNA (Cytosine-5-)-methyltransferase 1 (DNMT1) gene that causes hypermethylation of various tumor-suppressor genes and leads to a poor prognosis in GBM, was detected. Two hallmarks of cancer cells-proliferation and cell death-were positively influenced by riluzole treatment. Finally, we observed that riluzole reduced the tumor growth in in vivo CAM assay, suggesting it could be a possible synergistic drug for the treatment of glioblastoma.

17.
Arch Gen Psychiatry ; 68(12): 1247-56, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21810631

RESUMO

CONTEXT: Stress plays a major role in the development of comorbid alcohol use disorder (AUD). In turn, AUD worsens the outcome of psychiatric patients with respect to global disease severity, social situation, and socioeconomic burden. Prediction of persons at risk for AUD is crucial for future preventive and therapeutic strategies. OBJECTIVE: To investigate whether genetic variants of the corticotropin-releasing factor system or their interaction influence the risk of developing AUD in chronic disease populations. DESIGN: Genotype analysis comprising selected single-nucleotide polymorphisms within the CRHR1 and CRHBP genes in patients with schizophrenia and in a nonschizophrenic psychiatric disease control sample should allow the extraction of predictors of comorbid AUD. Gene expression (messenger RNA) analysis in peripheral blood mononuclear cells was performed to gain the first mechanistic insight. SETTING: An ideal setup for this study was the Göttingen Research Association for Schizophrenia Data Collection of schizophrenic patients, specifically intended to enable association of genetic information with quantifiable phenotypes in a phenotype-based genetic association study. Patients  A total of 1037 schizophrenic patients (Göttingen Research Association for Schizophrenia sample), 80 nonschizophrenic psychiatric disease controls as a small replicate sample, and a case-control study including 1141 healthy subjects. MAIN OUTCOME MEASURES: Association of CRHR1 and CRHBP genotypes with the following: (1) AUD; (2) a newly developed alcoholism severity score comprising 5 AUD-relevant variables; and (3) quantitative CRHR1 and CRHBP messenger RNA expression. RESULTS: An interaction of CRHR1 rs110402 and CRHBP rs3811939 predicts high risk of comorbid AUD in schizophrenic patients (odds ratio = 2.27; 95% confidence interval, 1.56-3.30; P < .001) as well as psychiatric disease controls (odds ratio = 4.02; 95% confidence interval, 0.95-17.05; P = .06) and leads to the highest CRHR1/CRHBP messenger RNA ratio (P = .02; dysbalanced stress axis). CONCLUSIONS: The high predictive value of a genetic interaction within the stress axis for the risk of comorbid AUD may be used for novel preventive and individualized therapeutic approaches.


Assuntos
Alcoolismo/genética , Proteínas de Transporte/genética , Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/genética , Esquizofrenia/genética , Adolescente , Adulto , Idoso , Alcoolismo/epidemiologia , Alcoolismo/fisiopatologia , Estudos de Casos e Controles , Comorbidade , Hormônio Liberador da Corticotropina/fisiologia , Estudos Transversais , Feminino , Estudos de Associação Genética , Humanos , Masculino , Transtornos Mentais/genética , Transtornos Mentais/fisiopatologia , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco , Esquizofrenia/epidemiologia , Esquizofrenia/fisiopatologia , Adulto Jovem
18.
Behav Brain Res ; 208(1): 80-4, 2010 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19900484

RESUMO

We have previously shown that high-dose erythropoietin (EPO) treatment improves hippocampal plasticity and cognitive performance in rodents and in patients suffering from neuropsychiatric diseases. It was therefore of interest to explore whether upregulation of endogenous EPO in brain by hypoxia inducible factor (HIF) stabilization would increase hippocampal memory similar to exogenous EPO. HIFs are transcription factors involved in the cellular response to low oxygen, including upregulation of transcripts like vascular endothelial growth factor (VEGF) and EPO. Under normal oxygen, prolylhydroxylases decrease HIF-alpha stability. This is banned by prolylhydroxylase inhibitors, which prevent oxygen dependent degradation and thus prolong HIF-alpha half life. In an experimental set-up identical to the one yielding strong cognitive effects with EPO, healthy male 28-day-old mice received FG-4497, a HIF prolylhydroxylase inhibitor, or placebo intraperitoneally every other day for 3 weeks. Behavioral testing and hematocrit determinations were conducted in independent cohorts at 1, 3, or 4 weeks after treatment completion. Increased EPO and VEGF mRNA expression in hippocampus or primary hippocampal neurons 6h after the application of FG-4497 confirmed its ability to stabilize HIF and upregulate HIF dependent transcription in brain. At 3 and 4 weeks after the last injection, respectively, FG-4497 treated mice compared to placebo mice had improved hippocampal memory in fear conditioning without change in hematocrit. In contrast, no improvement in memory was detected at 1 week, when the hematocrit was increased, indicating that cognitive improvement and hematocrit are not directly related. FG-4497 application for 3 weeks leads to delayed but lasting enhancement of hippocampal memory, making HIF stabilization an attractive target for pharmacological manipulation of cognition.


Assuntos
Hipocampo/fisiologia , Fator 1 Induzível por Hipóxia/metabolismo , Memória/efeitos dos fármacos , Estimulação Acústica/efeitos adversos , Análise de Variância , Animais , Animais Recém-Nascidos , Comportamento Animal , Células Cultivadas , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Inibidores Enzimáticos/farmacologia , Eritropoetina/genética , Eritropoetina/metabolismo , Medo , Hematócrito/métodos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Fator 1 Induzível por Hipóxia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Arch Gen Psychiatry ; 67(9): 879-88, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20819981

RESUMO

CONTEXT: Schizophrenia is the collective term for a heterogeneous group of mental disorders with a still obscure biological basis. In particular, the specific contribution of risk or candidate gene variants to the complex schizophrenic phenotype is largely unknown. OBJECTIVE: To prepare the ground for a novel "phenomics" approach, a unique schizophrenia patient database was established by GRAS (Göttingen Research Association for Schizophrenia), designed to allow association of genetic information with quantifiable phenotypes.Because synaptic dysfunction plays a key role in schizophrenia, the complexin 2 gene (CPLX2) was examined in the first phenotype-based genetic association study (PGAS) of GRAS [corrected] DESIGN: Subsequent to a classic case-control approach, we analyzed the contribution of CPLX2 polymorphisms to discrete cognitive domains within the schizophrenic population. To gain mechanistic insight into how certain CPLX2 variants influence gene expression and function, peripheral blood mononuclear cells of patients, Cplx -null mutant mice, and transfected cells were investigated. SETTING: Coordinating research center (Max Planck Institute of Experimental Medicine) and 23 collaborating psychiatric centers all over Germany. PARTICIPANTS: One thousand seventy-one patients with schizophrenia (DSM-IV) examined by an invariant investigator team, resulting in the GRAS database with more than 3000 phenotypic data points per patient, and 1079 healthy control subjects of comparable ethnicity. Main Outcome Measure Cognitive performance including executive functioning, reasoning, and verbal learning/memory. RESULTS: Six single-nucleotide polymorphisms, distributed over the whole CPLX2 gene, were found to be highly associated with current cognition of schizophrenic subjects but only marginally with premorbid intelligence. Correspondingly, in Cplx2 -null mutant mice, prominent cognitive loss of function was obtained only in combination with a minor brain lesion applied during puberty, modeling a clinically relevant environmental risk ("second hit") for schizophrenia. In the human CPLX2 gene, 1 of the identified 6 cognition-relevant single-nucleotide polymorphisms, rs3822674 in the 3' untranslated region, was detected to influence microRNA-498 binding and gene expression. The same marker was associated with differential expression of CPLX2 in peripheral blood mononuclear cells. CONCLUSIONS: The PGAS allows identification of marker-associated clinical/biological traits. Current cognitive performance in schizophrenic patients is modified by CPLX2 variants modulating posttranscriptional gene expression.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Transtornos Cognitivos/genética , Proteínas do Tecido Nervoso/genética , Polimorfismo de Nucleotídeo Único/genética , Esquizofrenia/genética , Adolescente , Adulto , Animais , Transtornos Cognitivos/diagnóstico , Bases de Dados Genéticas/estatística & dados numéricos , Feminino , Expressão Gênica/genética , Expressão Gênica/fisiologia , Estudos de Associação Genética , Marcadores Genéticos , Variação Genética/genética , Genótipo , Humanos , Masculino , Camundongos , Camundongos Mutantes , Pessoa de Meia-Idade , Testes Neuropsicológicos , Fenótipo , Psicologia do Esquizofrênico
20.
Mol Cancer Ther ; 8(8): 2452-60, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19671740

RESUMO

The endothelin (ET) axis, often deregulated in cancers, is a promising target for anticancer strategies. Whereas previous investigations have focused mostly on ET action in malignant cells, we chose a model allowing separate assessment of the effects of ETs and their receptors ET(A)R and ET(B)R in the tumor cells and the stromal compartment, which is increasingly recognized as a key player in cancer progression. In homozygous spotting lethal rats (sl/sl), a model of constitutive ET(B)R deficiency, we showed significant reduction of growth and metastasis of MAT B III rat mammary adenocarcinoma cells overexpressing ET(A)R and ET-1 but negative for ET(B)R. Lack of stromal ET(B)R expression did not influence angiogenesis. However, it was correlated with diminished infiltration by tumor-associated macrophages and with reduced production of tumor necrosis factor-alpha, both known as powerful promoters of tumor progression. These effects were almost completely abolished in transgenic sl/sl rats, wherein ET(B)R function is restored by expression of an intact ET(B)R transgene. This shows that tumor growth and metastasis are critically dependent on ET(B)R function in cells of the microenvironment and suggests that successful ETR antagonist therapy should also target the stromal component of ET signaling


Assuntos
Neoplasias Mamárias Experimentais/patologia , Receptor de Endotelina B/genética , Animais , Linhagem Celular Tumoral , Feminino , Metástase Neoplásica , Ratos , Ratos Transgênicos , Receptor de Endotelina B/metabolismo , Células Estromais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA