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











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 14062, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34234256

RESUMO

Medulloblastoma is the most common high-grade brain tumor in childhood. Medulloblastomas with c-myc amplification, classified as group 3, are the most aggressive among the four disease subtypes resulting in a 5-year overall survival of just above 50%. Despite current intensive therapy regimens, patients suffering from group 3 medulloblastoma urgently require new therapeutic options. Using a recently established c-myc amplified human medulloblastoma cell line, we performed an in-vitro-drug screen with single and combinatorial drugs that are either already clinically approved or agents in the advanced stage of clinical development. Candidate drugs were identified in vitro and then evaluated in vivo. Tumor growth was closely monitored by BLI. Vessel development was assessed by 3D light-sheet-fluorescence-microscopy. We identified the combination of gemcitabine and axitinib to be highly cytotoxic, requiring only low picomolar concentrations when used in combination. In the orthotopic model, gemcitabine and axitinib showed efficacy in terms of tumor control and survival. In both models, gemcitabine and axitinib were better tolerated than the standard regimen comprising of cisplatin and etoposide phosphate. 3D light-sheet-fluorescence-microscopy of intact tumors revealed thinning and rarefication of tumor vessels, providing one explanation for reduced tumor growth. Thus, the combination of the two drugs gemcitabine and axitinib has favorable effects on preventing tumor progression in an orthotopic group 3 medulloblastoma xenograft model while exhibiting a favorable toxicity profile. The combination merits further exploration as a new approach to treat high-risk group 3 medulloblastoma.


Assuntos
Antineoplásicos/farmacologia , Axitinibe/farmacologia , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Amplificação de Genes , Meduloblastoma/genética , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Desoxicitidina/farmacologia , Modelos Animais de Doenças , Expressão Gênica , Humanos , Meduloblastoma/tratamento farmacológico , Meduloblastoma/patologia , Camundongos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
2.
J Neurotrauma ; 37(2): 295-304, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31441378

RESUMO

The weight-drop model is used widely to replicate closed-head injuries in mice; however, the histopathological and functional outcomes may vary significantly between laboratories. Because skull fractures are reported to occur in this model, we aimed to evaluate whether these breaks may influence the variability of the weight-drop (WD) model. Male Swiss Webster mice underwent WD injury with either a 2 or 5 mm cone tip, and behavior was assessed at 2 h and 24 h thereafter using the neurological severity score. The expression of interleukin (IL)-6, IL-1ß, tumor necrosis factor-α, matrix metalloproteinase-9, and tissue inhibitor of metalloproteinase-1 genes was measured at 12 h and 1, 3, and 14 days after injury. Before the injury, micro-computed tomography (micro-CT) was performed to quantify skull thickness at the impact site. With a conventional tip diameter of 2 mm, 33% of mice showed fractures of the parietal bone; the 5 mm tip produced only 10% fractures. Compared with mice without fractures, mice with fractures had a severity-dependent worse functional outcome and a more pronounced upregulation of inflammatory genes in the brain. Older mice were associated with thicker parietal bones and were less prone to skull fractures. In addition, mice that underwent traumatic brain injury (TBI) with skull fracture had macroscopic brain damage because of skull depression. Skull fractures explain a considerable proportion of the variability observed in the WD model in mice-i.e., mice with skull fractures have a much stronger inflammatory response than do mice without fractures. Using older mice with thicker skull bones and an impact cone with a larger diameter reduces the rate of skull fractures and the variability in this very useful closed-head TBI model.


Assuntos
Lesões Encefálicas Traumáticas/etiologia , Modelos Animais de Doenças , Traumatismos Cranianos Fechados/complicações , Inflamação/etiologia , Fraturas Cranianas/etiologia , Animais , Masculino , Camundongos
3.
Mol Med ; 21: 185-96, 2015 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-25811991

RESUMO

Parent-of-origin imprints have been implicated in the regulation of neural differentiation and brain development. Previously we have shown that, despite the lack of a paternal genome, human parthenogenetic (PG) embryonic stem cells (hESCs) can form proliferating neural stem cells (NSCs) that are capable of differentiation into physiologically functional neurons while maintaining allele-specific expression of imprinted genes. Since biparental ("normal") hESC-derived NSCs (N NSCs) are targeted by immune cells, we characterized the immunogenicity of PG NSCs. Flow cytometry and immunocytochemistry revealed that both N NSCs and PG NSCs exhibited surface expression of human leukocyte antigen (HLA) class I but not HLA-DR molecules. Functional analyses using an in vitro mixed lymphocyte reaction assay resulted in less proliferation of peripheral blood mononuclear cells (PBMC) with PG compared with N NSCs. In addition, natural killer (NK) cells cytolyzed PG less than N NSCs. At a molecular level, expression analyses of immune regulatory factors revealed higher HLA-G levels in PG compared with N NSCs. In line with this finding, MIR152, which represses HLA-G expression, is less transcribed in PG compared with N cells. Blockage of HLA-G receptors ILT2 and KIR2DL4 on natural killer cell leukemia (NKL) cells increased cytolysis of PG NSCs. Together this indicates that PG NSCs have unique immunological properties due to elevated HLA-G expression.


Assuntos
Diferenciação Celular , Citotoxicidade Imunológica , Células-Tronco Embrionárias/citologia , Expressão Gênica , Antígenos HLA-G/genética , Células Matadoras Naturais/imunologia , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/metabolismo , Apoptose/genética , Apoptose/imunologia , Linhagem Celular , Regulação da Expressão Gênica , Antígenos HLA-DR/genética , Antígenos HLA-DR/imunologia , Antígenos HLA-DR/metabolismo , Antígenos HLA-G/imunologia , Antígenos HLA-G/metabolismo , Humanos , Células Matadoras Naturais/metabolismo , MicroRNAs/genética , Células-Tronco Neurais/citologia
4.
Cell Med ; 5(1): 29-42, 2013 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-26858862

RESUMO

Uniparental zygotes with two paternal (androgenetic, AG) or two maternal genomes (gynogenetic, GG) cannot develop into viable offsprings but form blastocysts from which pluripotent embryonic stem (ES) cells can be derived. For most organs, it is unclear whether uniparental ES cells can give rise to stably expandable somatic stem cells that can repair injured tissues. Even if previous reports indicated that the capacity of AG ES cells to differentiate in vitro into pan-neural progenitor cells (pNPCs) and into cells expressing neural markers is similar to biparental [normal fertilized (N)] ES cells, their potential for functional neurogenesis is not known. Here we show that murine AG pNPCs give rise to neuron-like cells, which then generate sodium-driven action potentials while maintaining fidelity of imprinted gene expression. Neural engraftment after intracerebral transplantation was achieved only by late (22 days) AG and N pNPCs with in vitro low colony-forming cell (CFC) capacity. However, persisting CFC formation seen, in particular, in early (13 or 16 days) differentiation cultures of N and AG pNPCs correlated with a high incidence of trigerm layer teratomas. As AG ES cells display functional neurogenesis and in vivo stability similar to N ES cells, they represent a unique model system to study the roles of paternal and maternal genomes on neural development and on the development of imprinting-associated brain diseases.

5.
J Cereb Blood Flow Metab ; 32(9): 1747-56, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22569191

RESUMO

The two bradykinin receptors B1R and B2R are central components of the kallikrein-kinin system with different expression kinetics and binding characteristics. Activation of these receptors by kinins triggers inflammatory responses in the target organ and in most situations enhances tissue damage. We could recently show that blocking of B1R, but not B2R, protects from cortical cryolesion by reducing inflammation and edema formation. In the present study, we investigated the role of B1R and B2R in a closed head model of focal traumatic brain injury (TBI; weight drop). Increased expression of B1R in the injured hemispheres of wild-type mice was restricted to the later stages after brain trauma, i.e. day 7 (P<0.05), whereas no significant induction could be observed for the B2R (P>0.05). Mice lacking the B1R, but not the B2R, showed less functional deficits on day 3 (P<0.001) and day 7 (P<0.001) compared with controls. Pharmacological blocking of B1R in wild-type mice had similar effects. Reduced axonal injury and astroglia activation could be identified as underlying mechanisms, while inhibition of B1R had only little influence on the local inflammatory response in this model. Inhibition of B1R may become a novel strategy to counteract trauma-induced neurodegeneration.


Assuntos
Astrócitos/efeitos dos fármacos , Axônios/patologia , Antagonistas de Receptor B1 da Bradicinina , Traumatismos Cranianos Fechados/tratamento farmacológico , Traumatismos Cranianos Fechados/patologia , Animais , Apoptose/efeitos dos fármacos , Comportamento Animal/fisiologia , Bradicinina/análogos & derivados , Bradicinina/farmacologia , Antagonistas de Receptor B2 da Bradicinina , Traumatismos Cranianos Fechados/metabolismo , Imuno-Histoquímica , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Receptor B1 da Bradicinina/biossíntese , Receptor B1 da Bradicinina/genética , Receptor B2 da Bradicinina/biossíntese , Receptor B2 da Bradicinina/genética , Recuperação de Função Fisiológica , Fator de Necrose Tumoral alfa/metabolismo
6.
Brain ; 133(11): 3166-80, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20884644

RESUMO

Synaptic inhibition is a central factor in the fine tuning of neuronal activity in the central nervous system. Symptoms consistent with reduced inhibition such as stiffness, spasms and anxiety occur in paraneoplastic stiff person syndrome with autoantibodies against the intracellular synaptic protein amphiphysin. Here we show that intrathecal application of purified anti-amphiphysin immunoglobulin G antibodies induces stiff person syndrome-like symptoms in rats, including stiffness and muscle spasms. Using in vivo recordings of Hoffmann reflexes and dorsal root potentials, we identified reduced presynaptic GABAergic inhibition as an underlying mechanism. Anti-amphiphysin immunoglobulin G was internalized into neurons by an epitope-specific mechanism and colocalized in vivo with presynaptic vesicular proteins, as shown by stimulation emission depletion microscopy. Neurons from amphiphysin deficient mice that did not internalize the immunoglobulin provided additional evidence of the specificity in antibody uptake. GABAergic synapses appeared more vulnerable than glutamatergic synapses to defective endocytosis induced by anti-amphiphysin immunoglobulin G, as shown by increased clustering of the endocytic protein AP180 and by defective loading of FM 1-43, a styryl dye used to label cell membranes. Incubation of cultured neurons with anti-amphiphysin immunoglobulin G reduced basal and stimulated release of γ-aminobutyric acid substantially more than that of glutamate. By whole-cell patch-clamp analysis of GABAergic inhibitory transmission in hippocampus granule cells we showed a faster, activity-dependent decrease of the amplitude of evoked inhibitory postsynaptic currents in brain slices treated with antibodies against amphiphysin. We suggest that these findings may explain the pathophysiology of the core signs of stiff person syndrome at the molecular level and show that autoantibodies can alter the function of inhibitory synapses in vivo upon binding to an intraneuronal key protein by disturbing vesicular endocytosis.


Assuntos
Autoanticorpos/uso terapêutico , Proteínas do Tecido Nervoso/imunologia , Inibição Neural/imunologia , Rigidez Muscular Espasmódica/imunologia , Rigidez Muscular Espasmódica/terapia , Ácido gama-Aminobutírico/metabolismo , Idoso , Animais , Autoanticorpos/administração & dosagem , Autoanticorpos/fisiologia , Células Cultivadas , Endocitose/imunologia , Feminino , Humanos , Imunização Passiva/métodos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/fisiologia , Imunoglobulina G/uso terapêutico , Potenciais Pós-Sinápticos Inibidores/fisiologia , Injeções Espinhais , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Ratos , Ratos Endogâmicos Lew , Rigidez Muscular Espasmódica/patologia , Ácido gama-Aminobutírico/deficiência
8.
Int J Dev Biol ; 54(11-12): 1755-62, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21404193

RESUMO

Patient derived stem cell-based therapies are considered a future treatment option for Parkinson´s disease, a chronic and progressive brain neurodegenerative disorder characterized by depletion of dopaminergic neurons in the basal ganglia. While many aspects of the in vitro and in vivo differentiation potential of uniparental parthenogenetic (PG) and gynogenetic (GG) embryonic stem (ES) cells of several species have been studied, the capacity of androgenetic (AG) ES cells to develop into neuronal subtypes remains unclear. Here, we investigated the potential of murine AG ES cells to undergo dopaminergic differentiation both via directed in vitro differentiation, and in vivo, in ES cell-chimeric E12.5 and E16.5 brains. We show that similar to normal (N; developed from a zygote with maternal and paternal genomes) ES cells, AG cells generated dopaminergic neurons in vitro and in E12.5 and E16.5 chimeric brains following blastocyst injection. Expression of brain-specific imprinted genes was maintained in AG and normal dopaminergic cell cultures. Our results indicate that AG ES cells have dopaminergic differentiation potential in vitro and in vivo. This contrasts with previous reports of limited neural in vivo differentiation of AG cells in later brain development, and suggests that AG ES cells could be therapeutically relevant for future cellular replacement strategies for brain disease.


Assuntos
Encéfalo/embriologia , Diferenciação Celular , Dopamina/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Genoma , Neurônios/citologia , Animais , Blastocisto , Encéfalo/citologia , Linhagem Celular , Quimera/embriologia , Quimera/genética , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante de Células-Tronco , Zigoto
9.
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
10.
Exp Transl Stroke Med ; 1: 4, 2009 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20142991

RESUMO

The tissue protective functions of the hematopoietic growth factor erythropoietin (EPO) are independent of its action on erythropoiesis. EPO and its receptors (EPOR) are expressed in multiple brain cells during brain development and upregulated in the adult brain after injury. Peripherally administered EPO crosses the blood-brain barrier and activates in the brain anti-apoptotic, anti-oxidant and anti-inflammatory signaling in neurons, glial and cerebrovascular endothelial cells and stimulates angiogenesis and neurogenesis. These mechanisms underlie its potent tissue protective effects in experimental models of stroke, cerebral hemorrhage, traumatic brain injury, neuroinflammatory and neurodegenerative disease. The preclinical data in support of the use of EPO in brain disease have already been translated to first clinical pilot studies with encouraging results with the use of EPO as a neuroprotective agent.

11.
Cell Signal ; 20(1): 154-62, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18006272

RESUMO

Thrombopoietin (TPO), a hematopoietic growth factor regulating platelet production, and its receptor (TPOR) were recently shown to be expressed in the brain where they exert proapoptotic activity. Here we used PC12 cells, an established model of neuronal differentiation, to investigate the effects of TPO on neuronal survival and differentiation. These cells expressed TPOR mRNA. TPO increased cell death in neuronally differentiated PC12 cells but had no effect in undifferentiated cells. Surprisingly, TPO inhibited nerve growth factor (NGF)-induced differentiation of PC12 cells in a dose- and time-dependent manner. This inhibition was dependent on the activity of Janus kinase-2 (JAK2). Using phospho-kinase arrays and Western blot we found downregulation of the NGF-stimulated phosphorylation of the extracellular signal-regulated kinase p42ERK by TPO with no effect on phosphorylation of Akt or stress kinases. NGF-induced phosphorylation of ERK-activating kinases, MEK1/2 and C-RAF was also reduced by TPO while NGF-induced RAS activation was not attenuated by TPO treatment. In contrast to its inhibitory effects on NGF signalling, TPO had no effect on epidermal growth factor (EGF)-stimulated ERK phosphorylation or proliferation of PC12 cells. Our data indicate that TPO via activation of its receptor-bound JAK2 delays the NGF-dependent acquisition of neuronal phenotype and decreases neuronal survival by suppressing NGF-induced ERK activity.


Assuntos
Diferenciação Celular/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Crescimento Neural/fisiologia , Neurônios/citologia , Transdução de Sinais , Trombopoetina/fisiologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Fator de Crescimento Epidérmico/fisiologia , MAP Quinase Quinase 1/metabolismo , Neurônios/fisiologia , Células PC12 , Proteínas Proto-Oncogênicas c-raf/metabolismo , Ratos , Proteínas ras/metabolismo
12.
Neurochem Res ; 31(10): 1219-30, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17021950

RESUMO

Correct timing and spatial location of growth factor expression is critical for undisturbed brain development and functioning. In terminally differentiated cells distinct biological responses to growth factors may depend on cell type specific activation of signalling cascades. We show that the hematopoietic growth factors thrombopoietin (TPO) and granulocyte colony-stimulating factor (GCSF) exert cell type specific effects on survival, proliferation and the degree of phosphorylation of Akt1, ERK1/2 and STAT3 in rat hippocampal neurons and cortical astrocytes. In neurons, TPO induced cell death and selectively activated ERK1/2. GCSF protected neurons from TPO- and hypoxia-induced cell death via selective activation of Akt1. In astrocytes, neither TPO nor GCSF had any effect on cell viability but inhibited proliferation. This effect was accompanied by activation of ERK1/2 and inhibition of STAT3 activity. A balance between growth factors, their receptors and signalling proteins may play an important role in regulation of neural cell survival.


Assuntos
Astrócitos/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Trombopoetina/farmacologia , Animais , Astrócitos/metabolismo , Western Blotting , Células Cultivadas , Imunofluorescência , Hipocampo/citologia , Hipocampo/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neurônios/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Fator de Transcrição STAT3/metabolismo
13.
J Neurosurg Anesthesiol ; 18(2): 132-8, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16628067

RESUMO

The discovery of the broad neuroprotective potential of erythropoietin (EPO), an endogenous hematopoietic growth factor, has opened new therapeutic avenues in the treatment of brain diseases. EPO expression in the brain is induced by hypoxia. Practically all brain cells are capable of production and release of EPO and expression of its receptor. EPO exerts multifaceted protective effects on brain cells. It protects neuronal cells from noxious stimuli such as hypoxia, excess glutamate, serum deprivation or kainic acid exposure in vitro by targeting a variety of mechanisms and involves neuronal, glial and endothelial cell functions. In rodent models of ischemic stroke, EPO reduces infarct volume and improves functional outcome, but beneficial effects have also been observed in animal models of subarachnoid hemorrhage, intracerebral hemorrhage, traumatic brain injury, and spinal cord injury. EPO has a convenient therapeutic window upon ischemic stroke and favorable pharmacokinetics. Results from first therapeutic trials in humans are promising, but will need to be validated in larger trials. The safety profile and effectiveness of EPO in a wide variety of neurologic disease models make EPO a candidate compound for a potential first-line therapeutic for neurologic emergencies.


Assuntos
Encefalopatias/tratamento farmacológico , Encefalopatias/fisiopatologia , Eritropoetina/fisiologia , Eritropoetina/uso terapêutico , Animais , Química Encefálica/fisiologia , Eritropoetina/biossíntese , Eritropoetina/farmacocinética , Humanos , Transtornos Mentais/tratamento farmacológico , Doenças do Sistema Nervoso/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Proteínas Recombinantes
14.
Anat Embryol (Berl) ; 210(3): 209-19, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16151855

RESUMO

Recombinant human erythropoietin (EPO) is neuroprotective in animal models of adult spinal cord injury, and reduces apoptosis in adult dorsal root ganglia after spinal nerve crush. The present work demonstrates that spinal cord and dorsal root ganglia share dynamic expression patterns of EPO and its receptor (EPOR) during development. C57Bl mice from embryonic days (E) 8 (E8) to E19 were studied. In spinal cord and dorsal root ganglia, EPOR expression in all precursor cells preceded the expression of EPO in subsets of neurons. On E11, EPO-immunoreactive spinal motoneurons and ganglionic sensory neurons resided adjacent to EPOR-expressing radial glial cells and satellite cells, respectively. From E12 onwards, EPOR-immunoreactivity decreased in radial glial cells and, transiently, in satellite cells. Simultaneously, large-scale apoptosis of motoneurons and sensory neurons started, and subsets of neurons were labelled by antibodies against EPOR. Viable neurons expressed EPO and EPOR. Up to E12.5, apoptotic cells were EPOR-immunopositive, but variably EPO-immunonegative or EPO-immunopositive. Thereafter, EPO-immunonegative and EPOR-immunopositive apoptotic cells predominated. Our findings suggest that EPO-mediated neuron-glial and, later, neuron-neuronal interactions promote the differentiation and/or the survival of subsets of neurons and glial cells in central as well as in peripheral parts of the embryonic nervous system. Correspondingly, expression of phospho-Akt-1/protein-kinase B extensively overlapped expression sites of EPO and EPOR, but was absent from apoptotic cells. Identified other sites of EPO and/or EPOR expression include radial glial cells that transform to astrocytes, cells of the floor plate and notochord as well as neural crest-derived boundary cap cells at motor exit points and cells of the primary sympathetic chain.


Assuntos
Desenvolvimento Embrionário/fisiologia , Eritropoetina/metabolismo , Gânglios Espinais/embriologia , Organogênese/fisiologia , Receptores da Eritropoetina/metabolismo , Medula Espinal/embriologia , Animais , Apoptose/fisiologia , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Gânglios Espinais/metabolismo , Idade Gestacional , Técnicas Imunoenzimáticas , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Medula Espinal/metabolismo
15.
J Cereb Blood Flow Metab ; 25(4): 427-30, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15689954

RESUMO

Gender differences in neuropsychiatric disease are recognized but not well understood. Investigating the survival of primary rat hippocampal neurons in culture, we found significant and inverted gender differences on normoxia versus hypoxia. Male cells were more resistant under normoxia but more vulnerable under hypoxia than female cells. Male vulnerability pattern was acquired in cells from neonatally testosterone-primed females. Estrogens, acting via membrane receptors, had a higher neuroprotective power in male neurons, explained at least in part by the pronounced increase in estrogen receptor beta/alpha ratio during hypoxia in male cells only.


Assuntos
Estradiol/farmacologia , Hipóxia Encefálica/patologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/efeitos dos fármacos , Feminino , Genótipo , Masculino , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Caracteres Sexuais , Testosterona/farmacologia
16.
Proc Natl Acad Sci U S A ; 102(3): 862-7, 2005 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-15642952

RESUMO

Central nervous and hematopoietic systems share developmental features. We report that thrombopoietin (TPO), a stimulator of platelet formation, acts in the brain as a counterpart of erythropoietin (EPO), a hematopoietic growth factor with neuroprotective properties. TPO is most prominent in postnatal brain, whereas EPO is abundant in embryonic brain and decreases postnatally. Upon hypoxia, EPO and its receptor are rapidly reexpressed, whereas neuronal TPO and its receptor are down-regulated. Unexpectedly, TPO is strongly proapoptotic in the brain, causing death of newly generated neurons through the Ras-extracellular signal-regulated kinase 1/2 pathway. This effect is not only inhibited by EPO but also by neurotrophins. We suggest that the proapoptotic function of TPO helps to select for neurons that have acquired target-derived neurotrophic support.


Assuntos
Apoptose , Química Encefálica , Encéfalo/citologia , Eritropoetina/fisiologia , Trombopoetina/fisiologia , Animais , Encéfalo/metabolismo , Regulação da Expressão Gênica , Fatores de Crescimento de Células Hematopoéticas/fisiologia , Hipóxia/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/farmacologia , Neurônios/citologia , Neurônios/metabolismo , Prosencéfalo/química , Prosencéfalo/citologia , Rombencéfalo/química , Rombencéfalo/citologia
17.
Brain Pathol ; 14(4): 455-6, 458-9, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15605994

RESUMO

May 2004: We present the case of a male newborn (38th week of gestation) with a 3-week history of a sonographically detected parietal mass of 5-cm diameter. The entire mass was removed at surgery. Surprisingly, microscopy revealed an intracerebral hemorrhage and nests of glycophorin-A immunoreactive blasts and nucleated erythrocytes in the surrounding parenchyma. The final diagnosis was chronic intracerebral hemorrhage of unknown etiology with reactive changes of the surrounding brain tissue and perifocal extramedullary erythropoiesis. This case is unique because, to date, intracranial extramedullary erythropoiesis has only been described in chronic subdural hemorrhage.


Assuntos
Doenças do Sistema Nervoso Central/patologia , Hemorragia Cerebral/patologia , Doenças do Sistema Nervoso Central/metabolismo , Hemorragia Cerebral/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Imuno-Histoquímica/métodos , Recém-Nascido , Imageamento por Ressonância Magnética/métodos , Masculino , Sinaptofisina/metabolismo
18.
Transfus Apher Sci ; 31(1): 39-44, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15294194

RESUMO

Erythropoietin, a hematopoietic growth-factor possessing manifold, potent neuroprotective properties, after multiple testing in cell culture and animal studies now gradually finds its way into clinical neuroscience. The first time this took place was in 1998 with a pilot study in stroke patients, the "Göttingen EPO-Stroke-Trial". This study was able to demonstrate that EPO is perfectly well tolerated and safe with this indication. Furthermore, the EPO-treated patients showed a significantly better outcome regarding their clinical progress as well as regarding the infarct size as observed by MRI, when compared to the placebo treated patients. At the moment a multicenter study is being carried out in Germany.


Assuntos
Anemia/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Eritropoetina/uso terapêutico , Hematínicos/uso terapêutico , Hipóxia Encefálica/tratamento farmacológico , Infarto Cerebral/tratamento farmacológico , Ensaios Clínicos como Assunto , Humanos , Projetos Piloto , Segurança , Resultado do Tratamento
19.
Invest Ophthalmol Vis Sci ; 45(5): 1514-22, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15111610

RESUMO

PURPOSE: Erythropoietin (EPO) modulates erythropoiesis by inhibiting apoptosis in erythrocyte progenitors. Recently, EPO has been shown to be protective in experimental models of mechanical trauma, neuroinflammation, cerebral and retinal ischemia, and even in a human stroke trial. However, little is known about EPO signal transduction in vivo and the usefulness of EPO in the prevention of the chronic, purely apoptotic neuronal cell death that contributes to vision loss in glaucoma and the progression of neurodegenerative diseases. METHODS: EPO's effects and signaling in the retinal ganglion cell axotomy paradigm were studied by Western blot analysis and immunohistochemistry, receptor expression was characterized in the retina before and after lesion. EPO was injected into the vitreous body to investigate neuroprotection of axotomized rat RGCs. Moreover, EPO's effects were studied in cultures of immunopurified retinal ganglion cells. Signal-transduction pathways transmitting neuroprotective EPO effects in vivo were characterized by the use of specific kinase inhibitors, immunohistochemistry, and Western blot analysis. RESULTS: EPO receptors (EPORs) were expressed on RGC somata and dendrites in vivo. EPOR expression did not significantly change after axotomy. Application of EPO prevented death of neurotrophic-factor-deprived immunopurified rat RGCs in vitro, rescued axotomized RGCs in vivo, and prevented caspase-3 activation. EPO-induced Akt phosphorylation and survival-promoting EPO effects were completely abolished by inhibition of PI-3-kinase. EPO neuroprotection followed a bell-shaped dose-response curve in vitro and in vivo, whereas toxic EPO effects were never observed, even at high concentrations. CONCLUSIONS: These data support a potential role for EPO as a therapeutic molecule against predominantly apoptotic neuronal cell death in the context of glaucoma or neurodegenerative diseases and delineate the PI-3-K/Akt pathway as the predominant mediator of EPO neuroprotection in this in vivo paradigm of neuronal cell death.


Assuntos
Apoptose/efeitos dos fármacos , Eritropoetina/farmacologia , Fármacos Neuroprotetores/farmacologia , Proteínas Serina-Treonina Quinases , Células Ganglionares da Retina/patologia , Animais , Axotomia , Western Blotting , Caspase 3 , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Nervo Óptico/cirurgia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Sprague-Dawley , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes , Células Ganglionares da Retina/metabolismo , Transdução de Sinais
20.
Glia ; 42(4): 390-7, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12730959

RESUMO

The astrocytic endothelin (ET) receptors, ET(A) and ET(B), modulate calcium signaling and the astrocytic gap junctional network. The nonselective ET receptor ligand ET-1 inhibits gap junction permeability, an effect that can be blocked by tolbutamide. This mechanism may play a role in pathophysiological conditions such as ischemic stroke, characterized by elevated tissue ET-1 levels and hypertrophic-appearing reactive astrocytes. Therefore, the effect of ET-1 on cellular protein content was investigated in confluent once-passaged rat astrocyte cultures under serum-free conditions, by the Lowry method. Gap junction permeability was determined by the dye transfer technique. ET-1 prevented the decrease in astrocytic protein content observed in controls. The effect of ET-1 on cellular protein content was most pronounced in cultures seeded at high density, but it was attenuated in ET(B)-deficient (sl/sl) astrocytes. This effect could be blocked by the nonselective ET antagonist LU 302872 (10 micro M), as well as by the protein synthesis inhibitor cycloheximide (10 micro M). This increase in astrocytic protein content was inhibited by the ATP-sensitive K(+) channel blocker tolbutamide, which also antagonized the ET-1-induced reduction of gap junction permeability and reversed the morphological changes observed in astrocytes upon ET-1 treatment. Cytosine arabinoside (10 micro M), a DNA synthesis blocker, inhibited the ET-1-induced BrdU uptake without affecting the ET-1-induced increase in astrocytic protein content. To conclude, ET-1 induces an increase in astrocytic protein content as well as changes in astrocyte morphology in vitro. This hypertrophic response involves uncoupling of the astrocytic gap junctional network and is not dependent on DNA synthesis.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Endotelina-1/farmacologia , Animais , Antimetabólitos/farmacocinética , Antimetabólitos Antineoplásicos/farmacologia , Astrócitos/citologia , Bromodesoxiuridina/farmacocinética , Células Cultivadas , Cicloeximida/farmacologia , Citarabina/farmacologia , Antagonistas dos Receptores de Endotelina , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/fisiologia , Hipoglicemiantes/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Propionatos/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Mutantes , Ratos Wistar , Receptores de Endotelina/metabolismo , Tolbutamida/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA