Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Tissue Res ; 378(1): 1-14, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30989398

RESUMO

Parkinson's disease (PD) is pathologically characterized by the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and alpha-synucleinopathy. We mimic the disease pathology with overexpression of either the human α-syn wildtype (α-syn-WT) or E46K mutant form (α-syn-E46K) in DA neurons of the SNpc in adult rats using AAV2/DJ as a viral vector for the first time. Transduction efficiency was compared to an equal virus titer expressing the green fluorescent protein (GFP). Motor skills of all animals were evaluated in the cylinder and amphetamine-induced rotation test over a total time period of 12 weeks. Additionally, stereological quantification of DA cells and striatal fiber density measurements were performed every 4 weeks after injection. Rats overexpressing α-syn-WT showed a progressive loss of DA neurons with 40% reduction after 12 weeks accompanied by a greater loss of striatal DA fibers. In contrast, α-syn-E46K led to this reduction after 4 weeks without further progress. Insoluble α-syn positive cytoplasmic inclusions were observed in both groups within DA neurons of the SNpc and VTA. In addition, both α-syn groups developed a characteristic worsening of the rotational behavior over time. However, only the α-syn-WT group reached statistically significant different values in the cylinder test. Summarizing these effects, we established a motor symptom animal model of PD by using AAV2/DJ in the brain for the first time. Thereby, overexpressing of α-syn-E46K mimicked a rather pre-symptomatic stage of the disease, while the α-syn-WT overexpressing animals imitated an early symptomatic stage of PD.


Assuntos
Modelos Animais de Doenças , Doença de Parkinson/metabolismo , Parte Compacta da Substância Negra/metabolismo , alfa-Sinucleína/metabolismo , Animais , Dependovirus , Feminino , Vetores Genéticos , Parvovirinae/genética , Ratos , Ratos Sprague-Dawley
2.
Neuroscience ; 360: 197-209, 2017 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-28790019

RESUMO

The loss of nigral dopaminergic neurons and the resulting dopamine (DA) depletion in the striatum (STR) lead to altered neuronal activity and enhanced beta activity in various regions of the basal ganglia (BG) motor loop in patients with Parkinson's disease and in rodents in the 6-hydroxydopamine (6-OHDA)-lesioned rat model. Intrastriatal DA graft implantation has been shown to re-innervate the host brain and restore DA input. Here, DA cell grafts were implanted into the STR of 6-OHDA-lesioned rats and the effect on neuronal activity under urethane anesthesia (1.4g/kg, injected intraperitoneally) was tested in the entopeduncular nucleus (EPN, the equivalent to the human globus pallidus internus), the output nucleus of the BG, and the globus pallidus (GP, the equivalent to the human globus pallidus externus), a key region in the indirect pathway. In animals, which were transplanted with cells derived from the ventral mesencephalon of embryonic day 12 rat embryos into the STR, the rotational behavior induced by DA agonists in 6-OHDA-lesioned rats was significantly improved. This was accompanied by alleviated EPN firing rate and reinstated patterns of neuronal activity in the GP and EPN. Analysis of oscillatory activity revealed enhanced beta activity in both regions, which was reduced after grafting. In summary these data indicate restoration of BG motor loop toward normal activity by DA graft integration.


Assuntos
Potenciais de Ação , Gânglios da Base , Neurônios Dopaminérgicos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Gânglios da Base/efeitos dos fármacos , Gânglios da Base/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Agonistas de Dopamina/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Feminino , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Oxidopamina/farmacologia , Doença de Parkinson/fisiopatologia , Ratos Sprague-Dawley , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo
3.
Exp Neurol ; 294: 19-31, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28445715

RESUMO

Several findings support the concept that sensorimotor integration is disturbed in Parkinson's disease (PD) and in levodopa-induced dyskinesias. In this study, we explored the neuronal firing activity of excitatory pyramidal cells and inhibitory interneurons in the forelimb region of the primary somatosensory cortex (S1FL-Ctx), along with its interaction with oscillatory activity of the primary motor cortex (MCtx) in 6-hydroxydopamine lesioned hemiparkinsonian (HP) and levodopa-primed dyskinetic (HP-LID) rats as compared to controls under urethane (1.4g/kg, i.p.) anesthesia. Further, gene expression patterns of distinct markers for inhibitory GABAergic neurons were analyzed in both cortical regions. While firing frequency and burst activity of S1FL-Ctx inhibitory interneurons were reduced in HP and HP-LID rats, measures of irregularity were enhanced in pyramidal cells. Further, enhanced coherence of distinct frequency bands of the theta/alpha, high-beta, and gamma frequency, together with enhanced synchronization of putative pyramidal cells and interneurons with MCtx oscillatory activity were observed. While GABA level was similar, gene expression levels of interneuron and GABAergic markers in S1FL-Ctx and MCtx of HP-LID rats differed to some extent. Our study shows that in a rat model of PD with dyskinesias, neuronal activity in putative interneurons was reduced, which was accompanied by high beta and gamma coherence between S1FL-Ctx and MCtx, together with changes in gene expression, indicating maladaptive neuroplasticity after long term levodopa treatment.


Assuntos
Potenciais de Ação/fisiologia , Discinesia Induzida por Medicamentos/patologia , Córtex Motor/patologia , Neurônios/fisiologia , Doença de Parkinson Secundária/patologia , Potenciais de Ação/efeitos dos fármacos , Animais , Antiparkinsonianos/efeitos adversos , Apomorfina/farmacologia , Modelos Animais de Doenças , Feminino , Glutamato Descarboxilase/metabolismo , Levodopa/efeitos adversos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Oxidopamina/toxicidade , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/tratamento farmacológico , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de GABA/genética , Receptores de GABA/metabolismo , Simpatolíticos/toxicidade , Espectrometria de Massas em Tandem
4.
Cell Transplant ; 24(8): 1451-67, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25199117

RESUMO

Substitution of degenerated dopaminergic (DA) neurons by intrastriatally transplanted ventral mesencephalon (VM)-derived progenitor cells has been shown to improve motor functions in parkinsonian patients and animal models, whereas investigations of electrophysiological properties of the grafted DA neurons have been rarely performed. Here we show electrophysiological properties of grafted VM progenitor cells at different time intervals up to 12 weeks after transplantation measured in acute brain slices using eGFP-Flag transfection to identify the graft. We were able to classify typical DA neurons according to the biphasic progression (voltage "sag") to hyperpolarizing current injections. Two types of DA-like neurons were classified. Whereas type 1 neurons were characterized by delayed action potentials after hyperpolarization and irregular spontaneous firing, type 2 neurons displayed burst firing after hyperpolarization, spontaneous bursts, and regular firing. Comparison to identified DA neurons in vitro indicates a high integration of the intrastriatally grafted neurons, since in vitro cultures displayed regular firing spontaneously, whereas grafted identified DA neurons showed irregular firing. Additionally, type 1 and type 2 neurons exhibited a slight increase in the spontaneous firing frequency over time intervals after grafting, which might reflect a progressive integration of the grafted DA neurons. Our results provide evidence of the differentiation of grafted VM progenitor cells into mature integrated DA neurons, which are shown to replace the missing DA neurons functionally early after grafting.


Assuntos
Proteínas de Fluorescência Verde/metabolismo , Mesencéfalo/citologia , Neurônios/fisiologia , Células-Tronco/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Encéfalo/fisiologia , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/genética , Injeções Espinhais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Atividade Motora/efeitos dos fármacos , Oxidopamina/farmacologia , Ratos , Ratos Sprague-Dawley , Transplante de Células-Tronco , Células-Tronco/citologia , Transplante Homólogo
5.
Cell Transplant ; 24(10): 2099-112, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25420114

RESUMO

Human induced pluripotent stem cells (hiPSCs) are promising sources for regenerative therapies like the replacement of dopaminergic neurons in Parkinson's disease. They offer an unlimited cell source that can be standardized and optimized to produce applicable cell populations to gain maximal functional recovery. In the present study, human cord blood-derived iPSCs (hCBiPSCs) were differentiated into dopaminergic neurons utilizing two different in vitro protocols for neural induction: (protocol I) by fibroblast growth factor (FGF-2) signaling, (protocol II) by bone morphogenetic protein (BMP)/transforming growth factor (TGF-ß) inhibition. After maturation, in vitro increased numbers of tyrosine hydroxylase (TH)-positive neurons (7.4% of total cells) were observed by protocol II compared to 3.5% in protocol I. Furthermore, 3 weeks after transplantation in hemiparkinsonian rats in vivo, a reduced number of undifferentiated proliferating cells was achieved with protocol II. In contrast, proliferation still occurred in protocol I-derived grafts, resulting in tumor-like growth in two out of four animals 3 weeks after transplantation. Protocol II, however, did not increase the number of TH(+) cells in the striatal grafts of hemiparkinsonian rats. In conclusion, BMP/TGF-ß inhibition was more effective than FGF-2 signaling with regard to dopaminergic induction of hCBiPSCs in vitro and prevented graft overgrowth in vivo.


Assuntos
Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Neurônios Dopaminérgicos/citologia , Sangue Fetal/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Transplante de Células-Tronco , Animais , Células Cultivadas , Corpo Estriado/citologia , Feminino , Humanos , Lentivirus/metabolismo , Neostriado/citologia , Ratos Sprague-Dawley , Transplante de Células-Tronco/métodos
6.
Int J Nanomedicine ; 9: 5289-306, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25484582

RESUMO

PURPOSE: Innovative nerve conduits for peripheral nerve reconstruction are needed in order to specifically support peripheral nerve regeneration (PNR) whenever nerve autotransplantation is not an option. Specific support of PNR could be achieved by neurotrophic factor delivery within the nerve conduits via nanotechnology or stem cell engineering and transplantation. METHODS: Here, we comparatively investigated the bioactivity of selected neurotrophic factors conjugated to iron oxide nanoparticles (np-NTFs) and of bone marrow-derived stem cells genetically engineered to overexpress those neurotrophic factors (NTF-BMSCs). The neurite outgrowth inductive activity was monitored in culture systems of adult and neonatal rat sensory dorsal root ganglion neurons as well as in the cell line from rat pheochromocytoma (PC-12) cell sympathetic culture model system. RESULTS: We demonstrate that np-NTFs reliably support numeric neurite outgrowth in all utilized culture models. In some aspects, especially with regard to their long-term bioactivity, np-NTFs are even superior to free NTFs. Engineered NTF-BMSCs proved to be less effective in induction of sensory neurite outgrowth but demonstrated an increased bioactivity in the PC-12 cell culture system. In contrast, primary nontransfected BMSCs were as effective as np-NTFs in sensory neurite induction and demonstrated an impairment of neuronal differentiation in the PC-12 cell system. CONCLUSION: Our results evidence that nanotechnology as used in our setup is superior over stem cell engineering when it comes to in vitro models for PNR. Furthermore, np-NTFs can easily be suspended in regenerative hydrogel matrix and could be delivered that way to nerve conduits for future in vivo studies and medical application.


Assuntos
Engenharia Celular/métodos , Nanopartículas de Magnetita/química , Nanotecnologia/métodos , Fatores de Crescimento Neural/metabolismo , Neuritos/fisiologia , Animais , Células-Tronco Mesenquimais/citologia , Fatores de Crescimento Neural/genética , Regeneração Nervosa , Células PC12 , Ratos , Ratos Wistar , Medicina Regenerativa
7.
Behav Brain Res ; 261: 97-105, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24342749

RESUMO

Tactile stimulation (TS) applied to adult rats after cortical injury (medial frontal cortex aspiration or sensorimotor pial stripping stroke model) has been previously shown to ameliorate behavioral impairments and to improve morphological parameters like dendritic length of prefrontal cortical neurons (Gibb et al., 2010). The purpose of this study was to examine the effect of TS on healthy and hemiparkinsonian adult rats. Therefore, the animals received TS for 14 days and 15 min three times daily. At different time points rats were tested in various behavioral tests (amphetamine-induced rotation, cylinder test, staircase test). Finally, rats were sacrificed, their brains removed, and processed for Golgi-Cox analyses, tyrosine hydroxylase immunohistochemistry and quantitative RT-PCR. We found that the striatal 6-OHDA lesion itself induced a long-term increase of astroglial Fgf2 transcript levels, but was not further increased by TS. In contrast TS applied to healthy rats elicited a transient short-term increase of Fgf2 in the striatum and Bdnf, Grin1, and Fgf2 in the hippocampus. Moreover, behavioral and histological analyses do not support a beneficial effect of TS for hemiparkinsonian rats, applied for two weeks starting one day after partial striatal 6-OHDA lesion.


Assuntos
Lateralidade Funcional/fisiologia , Regulação da Expressão Gênica/fisiologia , Transtornos Parkinsonianos/reabilitação , Tato/fisiologia , Adrenérgicos/toxicidade , Anfetamina , Animais , Corpo Estriado/metabolismo , Dendritos/patologia , Dendritos/ultraestrutura , Modelos Animais de Doenças , Hipocampo/metabolismo , Masculino , Neurônios/metabolismo , Neurônios/patologia , Oxidopamina/toxicidade , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/complicações , Transtornos Parkinsonianos/patologia , Estimulação Física , Córtex Pré-Frontal/lesões , Córtex Pré-Frontal/patologia , Córtex Pré-Frontal/ultraestrutura , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Long-Evans , Fatores de Tempo
8.
PLoS One ; 8(12): e82871, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24358231

RESUMO

Nuclear bodies are large sub-nuclear structures composed of RNA and protein molecules. The Survival of Motor Neuron (SMN) protein localizes to Cajal bodies (CBs) and nuclear gems. Diminished cellular concentration of SMN is associated with the neurodegenerative disease Spinal Muscular Atrophy (SMA). How nuclear body architecture and its structural components influence neuronal differentiation remains elusive. In this study, we analyzed the effects of SMN and two of its interaction partners in cellular models of neuronal differentiation. The nuclear 23 kDa isoform of Fibroblast Growth Factor - 2 (FGF-2(23)) is one of these interacting proteins - and was previously observed to influence nuclear bodies by destabilizing nuclear gems and mobilizing SMN from Cajal bodies (CBs). Here we demonstrate that FGF-2(23) blocks SMN-promoted neurite outgrowth, and also show that SMN disrupts FGF-2(23)-dependent transcription. Our results indicate that FGF-2(23) and SMN form an inactive complex that interferes with neuronal differentiation by mutually antagonizing nuclear functions. Coilin is another nuclear SMN binding partner and a marker protein for Cajal bodies (CBs). In addition, coilin is essential for CB function in maturation of small nuclear ribonucleoprotein particles (snRNPs). The role of coilin outside of Cajal bodies and its putative impacts in tissue differentiation are poorly defined. The present study shows that protein levels of nucleoplasmic coilin outside of CBs decrease during neuronal differentiation. Overexpression of coilin has an inhibitory effect on neurite outgrowth. Furthermore, we find that nucleoplasmic coilin inhibits neurite outgrowth independent of SMN binding revealing a new function for coilin in neuronal differentiation.


Assuntos
Diferenciação Celular/genética , Núcleo Celular/metabolismo , Corpos Enovelados/metabolismo , Neurogênese/genética , Neurônios/fisiologia , Proteínas do Complexo SMN/fisiologia , Animais , Células Cultivadas , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Neuritos/fisiologia , Proteínas Nucleares/metabolismo , Células PC12 , Ligação Proteica , Ratos , Proteínas do Complexo SMN/metabolismo
9.
Biomaterials ; 34(38): 9886-904, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24050875

RESUMO

Biosynthetic nerve grafts are desired as alternative to autologous nerve grafts in peripheral nerve reconstruction. Artificial nerve conduits still have their limitations and are not widely accepted in the clinical setting. Here we report an analysis of fine-tuned chitosan tubes used to reconstruct 10 mm nerve defects in the adult rat. The chitosan tubes displayed low, medium and high degrees of acetylation (DAI: ≈ 2%, DA: ≈ 5%, DAIII: ≈ 20%) and therefore different degradability and microenvironments for the regenerating nerve tissue. Short and long term investigations were performed demonstrating that the chitosan tubes allowed functional and morphological nerve regeneration similar to autologous nerve grafts. Irrespective of the DA growth factor regulation demonstrated to be the same as in controls. Analyses of stereological parameters as well as the immunological tissue response at the implantation site and in the regenerated nerves, revealed that DAI and DAIII chitosan tubes displayed some limitations in the support of axonal regeneration and a high speed of degradation accompanied with low mechanical stability, respectively. The chitosan tubes combine several pre-requisites for a clinical acceptance and DAII chitosan tubes have to be judged as the most supportive for peripheral nerve regeneration.


Assuntos
Quitosana/química , Acetilação , Animais , Western Blotting , Cromatografia em Gel , Eletrofisiologia , Feminino , Regeneração Tecidual Guiada/métodos , Imuno-Histoquímica , Espectroscopia de Ressonância Magnética , Regeneração Nervosa/fisiologia , Nervos Periféricos/patologia , Nervos Periféricos/fisiologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Neurobiol Dis ; 59: 230-43, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23938762

RESUMO

Dopamine (DA) depletion in the nigrostriatal system leads to basal ganglia dysfunction both in Parkinson's disease (PD) and in 6-hydroxy dopamine (6-OHDA)-lesioned rats with neuronal hyperactivity in the subthalamic nucleus (STN), i.e. increased firing rate and burst activity, together with enhanced beta oscillatory activity. Moreover, intrastriatal transplantation of DA neurons has been shown to functionally re-innervate the host striatum and restore DA input. However, the effects of those transplanted cells on the STN are not well characterized. Therefore, we transplanted cells, derived from the ventral mesencephalon of E12 rat embryos, intrastriatally in the unilateral 6-OHDA-lesioned rat model of PD. We combined behavioral and histological findings with electrophysiological extracellular recordings in the STN, as well as qRT-PCR analyses of dopaminergic, GABAergic, and glutamatergic transporter and receptor genes in the striatum and the STN. Transplanted animals displayed improved rotational behavior after amphetamine injection by 50% in rats with small grafts (586±109 SEM dopamine cells), or even overcompensation by 116% in rats with large grafts (3486±548 SEM dopamine cells). Electrophysiological measurements revealed, that in rats with large grafts burst activity was not affected, while STN neuronal firing rate, as well as beta oscillatory activity was alleviated, whereas small grafts had less impact. Interestingly, both behavioral and electrophysiological measures were dependent on the number of surviving tyrosine hydroxylase positive cells. Although grafted rats displayed restored expression of the GABA synthesizing enzymes Gad65 and Gad67 in the striatum compared to naive rats, the grafts induced a decreased mRNA expression of dopamine receptor Drd2, glutamate receptors AMPA3, NMDA2A, and NMDA2B, and glutamate transporter Eaat3. Interestingly, the NMDA receptor subunit 2B and glutamate transporter Eaat3 were also less expressed in the STN of grafted animals compared to naive rats. In summary, DA grafts restore functional deficits and cause partial improvement of subthalamic neuronal activity. Incomplete recovery, however, may be due to decreased receptor gene expression induced by DA grafts in the striatum and in the STN.


Assuntos
Lateralidade Funcional/fisiologia , Regulação da Expressão Gênica/fisiologia , Neurônios/fisiologia , Doença de Parkinson/cirurgia , Núcleo Subtalâmico/metabolismo , Núcleo Subtalâmico/patologia , Potenciais de Ação/fisiologia , Adrenérgicos/toxicidade , Animais , Células Cultivadas , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Mesencéfalo/citologia , Oxidopamina/toxicidade , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Ratos , Ratos Sprague-Dawley , Transplante de Células-Tronco , Células-Tronco/fisiologia , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
11.
Cell Mol Life Sci ; 70(14): 2555-68, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23334184

RESUMO

Reduced levels of survival of motoneuron (SMN) protein lead to spinal muscular atrophy, but it is still unknown how SMN protects motoneurons in the spinal cord against degeneration. In the nucleus, SMN is associated with two types of nuclear bodies denoted as gems and Cajal bodies (CBs). The 23 kDa isoform of fibroblast growth factor-2 (FGF-2(23)) is a nuclear protein that binds to SMN and destabilizes the SMN-Gemin2 complex. In the present study, we show that FGF-2(23) depletes SMN from CBs without affecting their general structure. FRAP analysis of SMN-EGFP in CBs demonstrated that the majority of SMN in CBs remained mobile and allowed quantification of fast, slow and immobile nuclear SMN populations. The potential for SMN release was confirmed by in vivo photoconversion of SMN-Dendra2, indicating that CBs concentrate immobile SMN that could have a specialized function in CBs. FGF-2(23) accelerated SMN release from CBs, accompanied by a conversion of immobile SMN into a mobile population. Furthermore, FGF-2(23) caused snRNP accumulation in CBs. We propose a model in which Cajal bodies store immobile SMN that can be mobilized by its nuclear interaction partner FGF-2(23), leading to U4 snRNP accumulation in CBs, indicating a role for immobile SMN in tri-snRNP assembly.


Assuntos
Corpos Enovelados/metabolismo , Proteínas do Complexo SMN/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Células HEK293 , Humanos , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Proteínas Nucleares/metabolismo , Ligação Proteica , Mapas de Interação de Proteínas , Isoformas de Proteínas/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/metabolismo
12.
J Comp Neurol ; 520(17): 3949-61, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22592787

RESUMO

Fibroblast growth factor 2 (FGF-2) is an important neurotrophic factor that promotes survival of adult mesencephalic dopaminergic (mDA) neurons and regulates their adequate development. Since mDA neurons degenerate in Parkinson's disease, a comprehensive understanding of their development and maintenance might contribute to the development of causative therapeutic approaches. The current analysis addressed the role of FGF-2 in mDA axonal outgrowth, pathway formation, and innervation of respective forebrain targets using organotypic explant cocultures of ventral midbrain (VM) and forebrain (FB). An enhanced green fluorescent protein (EGFP) transgenic mouse strain was used for the VM explants, which allowed combining and distinguishing of individual VM and FB tissue from wildtype and FGF-2-deficient embryonic day (E)14.5 embryos, respectively. These cocultures provided a suitable model to study the role of target-derived FB and intrinsic VM-derived FGF-2. In fact, we show that loss of FGF-2 in both FB and VM results in significantly increased mDA fiber outgrowth compared to wildtype cocultures, proving a regulatory role of FGF-2 during nigrostriatal wiring. Further, we found in heterogeneous cocultures deficient for FGF-2 in FB and VM, respectively, similar phenotypes with wider fiber tracts compared to wildtype cocultures and shorter fiber outgrowth distance than cocultures completely deficient for FGF-2. Additionally, the loss of target-derived FGF-2 in FB explants resulted in decreased caudorostral glial migration. Together these findings imply an intricate interplay of target-derived and VM-derived FGF signaling, which assures an adequate nigrostriatal pathway formation and target innervation.


Assuntos
Corpo Estriado/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Vias Neurais/metabolismo , Neurogênese/fisiologia , Substância Negra/metabolismo , Animais , Técnicas de Cocultura , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
13.
Neurobiol Dis ; 47(2): 248-57, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22542539

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by selective motoneuron loss in brain and spinal cord. Mutations in the superoxide dismutase (SOD) 1 gene account for 10-20% of familial ALS patients. The ALS-mouse model over-expressing a mutant human SOD1 (G93A) gene closely mimics human ALS disease. The cause for the selective death of motoneurons is still unclear, but among several pathomechanisms discussed, loss of neurotrophic factors is one possibility. Basic fibroblast growth factor 2 (FGF-2) plays a prominent role in the motor system. In order to evaluate a role of FGF-2 in ALS pathogenesis, double mouse mutants transgenic for the human SOD1 mutation and lacking the endogenous FGF-2 gene were generated. Both heterozygous and homozygous FGF-2 deficient mutant SOD1 mice showed a significant delay in disease onset and less impaired motor performance in comparison to mutant SOD1 mice with normal FGF-2 levels. Survival of the double mouse mutants was significantly prolonged for two weeks. Motoneuron numbers were significantly higher in the double mutants and astrocytosis was diminished at disease endstage. While one would initially have expected that FGF-2 deficiency deteriorates the phenotype of mutant SOD1 animals, our results revealed a protective effect of FGF-2 reduction. In search of the underlying mechanisms, we could show up-regulation of other neurotrophic factors with proven protective effects in the ALS mouse model, ciliary neurotrophic factor (CNTF) and glial derived neurotrophic factor (GDNF) in muscle and spinal cord tissue of double mutant animals.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Modelos Animais de Doenças , Fator 2 de Crescimento de Fibroblastos/deficiência , Desempenho Psicomotor/fisiologia , Superóxido Dismutase/biossíntese , Fatores Etários , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Destreza Motora/fisiologia , Superóxido Dismutase-1 , Taxa de Sobrevida/tendências
14.
J Neurochem ; 122(1): 94-105, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22537018

RESUMO

Fibroblast growth factor 2 (FGF-2) is a neurotrophic factor participating in regulation of proliferation, differentiation, apoptosis and neuroprotection in the central nervous system. With regard to dopaminergic (DA) neurons of substantia nigra pars compacta (SNpc), which degenerate in Parkinson's disease, FGF-2 improves survival of mature DA neurons in vivo and regulates expansion of DA progenitors in vitro. To address the physiological role of FGF-2 in SNpc development, embryonic (E14.5), newborn (P0) and juvenile (P28) FGF-2-deficient mice were investigated. Stereological quantification of DA neurons identified normal numbers in the ventral tegmental area, whereas the SNpc of FGF-2-deficient mice displayed a 35% increase of DA neurons at P0 and P28, but not at earlier stage E14.5. Examination of DA marker gene expression by quantitative RT-PCR and in situ hybridization revealed a normal patterning of embryonic ventral mesencephalon. However, an increase of proliferating Lmx1a DA progenitors in the subventricular zone of the ventral mesencephalon of FGF-2-deficient embryos indicated altered cell cycle progression of neuronal progenitors. Increased levels of nuclear FgfR1 in E14.5 FGF-2-deficient mice suggest alterations of integrative nuclear FgfR1 signaling (INFS). In summary, FGF-2 restricts SNpc DA neurogenesis in vivo during late stages of embryonic development.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Substância Negra/citologia , Área Tegmentar Ventral/citologia , Fatores Etários , Animais , Animais Recém-Nascidos , Padronização Corporal/genética , Bromodesoxiuridina , Contagem de Células , Morte Celular/genética , Embrião de Mamíferos , Fator 2 de Crescimento de Fibroblastos/deficiência , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/genética , Substância Negra/embriologia , Substância Negra/crescimento & desenvolvimento , Fatores de Transcrição/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Área Tegmentar Ventral/embriologia , Área Tegmentar Ventral/crescimento & desenvolvimento
15.
J Cell Biochem ; 113(9): 2920-36, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22539306

RESUMO

FGF Receptor-1 (FGFR1), a membrane-targeted protein, is also involved in independent direct nuclear signaling. We show that nuclear accumulation of FGFR1 is a common response to retinoic acid (RA) in pluripotent embryonic stem cells (ESC) and neural progenitors and is both necessary and sufficient for neuronal-like differentiation and accompanying neuritic outgrowth. Dominant negative nuclear FGFR1, which lacks the tyrosine kinase domain, prevents RA-induced differentiation while full-length nuclear FGFR1 elicits differentiation in the absence of RA. Immunoprecipitation and GST assays demonstrate that FGFR1 interacts with RXR, RAR and their Nur77 and Nurr1 partners. Conditions that promote these interactions decrease the mobility of nuclear FGFR1 and RXR in live cells. RXR and FGFR1 co-associate with 5'-Fluorouridine-labeled transcription sites and with RA Responsive Elements (RARE). RA activation of neuronal (tyrosine hydroxylase) and neurogenic (fgf-2 and fgfr1) genes is accompanied by increased FGFR1, Nur, and histone H3.3 binding to their regulatory sequences. Reporter-gene assays show synergistic activations of RARE, NBRE, and NurRE by FGFR1, RAR/RXR, and Nurs. As shown for mESC differentiation, FGFR1 mediates gene activation by RA and augments transcription in the absence of RA. Cooperation of FGFR1 with RXR/RAR and Nurs at targeted genomic sequences offers a new mechanism in developmental gene regulation.


Assuntos
Células-Tronco Embrionárias/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores do Ácido Retinoico/metabolismo , Western Blotting , Células Cultivadas , Imunoprecipitação da Cromatina , Células-Tronco Embrionárias/citologia , Recuperação de Fluorescência Após Fotodegradação , Humanos , Imuno-Histoquímica , Imunoprecipitação , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptores do Ácido Retinoico/genética
16.
J Biol Chem ; 287(24): 19827-40, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22514272

RESUMO

Experiments in mice deficient for Nurr1 or expressing the dominant-negative FGF receptor (FGFR) identified orphan nuclear receptor Nurr1 and FGFR1 as essential factors in development of mesencephalic dopaminergic (mDA) neurons. FGFR1 affects brain cell development by two distinct mechanisms. Activation of cell surface FGFR1 by secreted FGFs stimulates proliferation of neural progenitor cells, whereas direct integrative nuclear FGFR1 signaling (INFS) is associated with an exit from the cell cycle and neuronal differentiation. Both Nurr1 and INFS activate expression of neuronal genes, such as tyrosine hydroxylase (TH), which is the rate-limiting enzyme in dopamine synthesis. Here, we show that nuclear FGFR1 and Nurr1 are expressed in the nuclei of developing TH-positive cells in the embryonic ventral midbrain. Both nuclear receptors were effectively co-immunoprecipitated from the ventral midbrain of FGF-2-deficient embryonic mice, which previously showed an increase of mDA neurons and enhanced nuclear FGFR1 accumulation. Immunoprecipitation and co-localization experiments showed the presence of Nurr1 and FGFR1 in common nuclear protein complexes. Fluorescence recovery after photobleaching and chromatin immunoprecipitation experiments demonstrated the Nurr1-mediated shift of nuclear FGFR1-EGFP mobility toward a transcriptionally active population and that both Nurr1 and FGFR1 bind to a common region in the TH gene promoter. Furthermore, nuclear FGFR1 or its 23-kDa FGF-2 ligand (FGF-2(23)) enhances Nurr1-dependent activation of the TH gene promoter. Transcriptional cooperation of FGFR1 with Nurr1 was confirmed on isolated Nurr1-binding elements. The proposed INFS/Nurr1 nuclear partnership provides a novel mechanism for TH gene regulation in mDA neurons and a potential therapeutic target in neurodevelopmental and neurodegenerative disorders.


Assuntos
Núcleo Celular/metabolismo , Neurônios Dopaminérgicos/metabolismo , Mesencéfalo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Núcleo Celular/genética , Neurônios Dopaminérgicos/citologia , Mesencéfalo/citologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Elementos de Resposta/fisiologia , Transcrição Gênica/fisiologia , Tirosina 3-Mono-Oxigenase/biossíntese
17.
PLoS One ; 7(2): e31202, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22348054

RESUMO

The monogenetic disease Spinal Muscular Atrophy (SMA) is characterized by a progressive loss of motoneurons leading to muscle weakness and atrophy due to severe reduction of the Survival of Motoneuron (SMN) protein. Several models of SMA show deficits in neurite outgrowth and maintenance of neuromuscular junction (NMJ) structure. Survival of motoneurons, axonal outgrowth and formation of NMJ is controlled by neurotrophic factors such as the Fibroblast Growth Factor (FGF) system. Besides their classical role as extracellular ligands, some FGFs exert also intracellular functions controlling neuronal differentiation. We have previously shown that intracellular FGF-2 binds to SMN and regulates the number of a subtype of nuclear bodies which are reduced in SMA patients. In the light of these findings, we systematically analyzed the FGF-system comprising five canonical receptors and 22 ligands in a severe mouse model of SMA. In this study, we demonstrate widespread alterations of the FGF-system in both muscle and spinal cord. Importantly, FGF-receptor 1 is upregulated in spinal cord at a pre-symptomatic stage as well as in a mouse motoneuron-like cell-line NSC34 based model of SMA. Consistent with that, phosphorylations of FGFR-downstream targets Akt and ERK are increased. Moreover, ERK hyper-phosphorylation is functionally linked to FGFR-1 as revealed by receptor inhibition experiments. Our study shows that the FGF system is dysregulated at an early stage in SMA and may contribute to the SMA pathogenesis.


Assuntos
Fatores de Crescimento de Fibroblastos/análise , Atrofia Muscular Espinal/etiologia , Proteínas do Complexo SMN/análise , Animais , Modelos Animais de Doenças , Sistema de Sinalização das MAP Quinases , Camundongos , Músculos/química , Atrofia Muscular Espinal/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/análise , Medula Espinal/química
18.
Cell Transplant ; 21(4): 749-62, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21929871

RESUMO

Exogenous cell replacement represents a potent treatment option for Parkinson's disease. However, the low survival rate of transplanted dopaminergic neurons (DA) calls for methodological improvements. Here we evaluated a method to combine transient genetic modification of neuronal progenitor cells with an optimized cell culture protocol prior to intrastriatal transplantation into 6-hydroxydopamine (6-OHDA) unilateral lesioned rats. Plasmid-based delivery of brain-derived neurotrophic factor (BDNF) increases the number of DA neurons, identified by tyrosine hydroxylase immunoreactivity (TH-ir), by 25% in vitro, compared to enhanced green fluorescence protein (EGFP)-transfected controls. However, the nucleofection itself, especially the cell detachment and reseeding procedure, decreases the TH-ir neuron number to 40% compared with nontransfected control cultures. To circumvent this drawback we established the colayer method, which contains a mix of nucleofected cells reseeded on top of an adherent sister culture in a ratio 1:3. In this setup TH-ir neuron number remains high and could be further increased by 25% after BDNF transfection. Comparison of both cell culture procedures (standard and colayer) after intrastriatal transplantation revealed a similar DA neuron survival as seen in vitro. Two weeks after grafting TH-ir neuron number was strongly reduced in animals receiving the standard EGFP-transfected cells (271 ± 62) compared to 1,723 ± 199 TH-ir neurons in the colayer group. In contrast to the in vitro results, no differences in the number of grafted TH-ir neurons were observed between BDNF, EGFP, and nontransfected colayer groups, neither 2 nor 13 weeks after transplantation. Likewise, amphetamine and apomorphine-induced rotational behavior improved similarly over time in all groups. Nevertheless, the colayer protocol provides an efficient way for neurotrophic factor release by transplanted progenitor cells and will help to study the effects of candidate factors on survival and integration of transplanted DA neurons.


Assuntos
Mesencéfalo/citologia , Oxidopamina/efeitos adversos , Doença de Parkinson/terapia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Animais , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Ratos , Ratos Sprague-Dawley , Células-Tronco/metabolismo
19.
PLoS One ; 6(8): e23564, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21876757

RESUMO

Secreted proteins of the fibroblast growth factor (FGF) family play important roles during development of various organ systems. A detailed knowledge of their temporal and spatial expression profiles, especially of closely related FGF family members, are essential to further identification of specific functions in distinct tissues. In the central nervous system dopaminergic neurons of the substantia nigra and their axonal projections into the striatum progressively degenerate in Parkinson's disease. In contrast, FGF-2 deficient mice display increased numbers of dopaminergic neurons. In this study, we determined the expression profiles of all 22 FGF-ligands and 10 FGF-receptor isoforms, in order to clarify, if FGF-2 deficiency leads to compensatory up-regulation of other FGFs in the nigrostriatal system. Three tissues, ventral mesencephalon (VM), striatum (STR) and as reference tissue spinal cord (SC) of wild-type and FGF-2 deficient mice at four developmental stages E14.5, P0, P28, and adult were comparatively analyzed by quantitative RT-PCR. As no differences between the genotypes were observed, a compensatory up-regulation can be excluded. Moreover, this analysis revealed that the majority of FGF-ligands (18/22) and FGF-receptors (9/10) are expressed during normal development of the nigrostriatal system and identified dynamic changes for some family members. By comparing relative expression level changes to SC reference tissue, general alterations in all 3 tissues, such as increased expression of FGF-1, -2, -22, FgfR-2c, -3c and decreased expression of FGF-13 during postnatal development were identified. Further, specific changes affecting only one tissue, such as increased FGF-16 (STR) or decreased FGF-17 (VM) expression, or two tissues, such as decreased expression of FGF-8 (VM, STR) and FGF-15 (SC, VM) were found. Moreover, 3 developmentally down-regulated FGFs (FGF-8b, FGF-15, FGF-17a) were functionally characterized by plasmid-based over-expression in dissociated E11.5 VM cell cultures, however, such a continuous exposure had no influence on the yield of dopaminergic neurons in vitro.


Assuntos
Fator 2 de Crescimento de Fibroblastos/deficiência , Regulação da Expressão Gênica no Desenvolvimento , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Substância Negra/crescimento & desenvolvimento , Substância Negra/metabolismo , Animais , Bases de Dados Genéticas , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Hibridização In Situ , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Fatores de Crescimento de Fibroblastos/genética
20.
J Neurotrauma ; 28(4): 661-74, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21265597

RESUMO

Short-term low-frequency electrical stimulation (ESTIM) of proximal peripheral nerve stumps prior to end-to-end coaptation or tubular bridging of small distances has been reported to increase preferential motor reinnervation and functional motor recovery in animal models and human patients undergoing carpal tunnel release surgery. We investigated the effects of ESTIM on regeneration across rat sciatic nerve gaps, which exceed distances that allow spontaneous regeneration. Three different reconstruction approaches were combined with ESTIM in the experimental groups. Nerve gaps (13 mm) were bridged using (I) nerve autotransplantation, (II) transplantation of differentially filled silicone tubes, or (III) transplantation of tubular grafts containing fibroblast growth factor-2 overexpressing Schwann cells (SCs) for gene therapy. The regeneration outcome was followed for up to 8 weeks, and functionally as well as histomorphometrically analyzed in comparison to non-stimulated control groups. Combining ESTIM with nerve autotransplantation significantly increased the nerve fiber density in the regenerated nerve, and the grade of functional recovery as detected by electrodiagnostic recordings from the gastrocnemius muscle. The combination of ESTIM with transplantation of naïve SCs increased the regeneration of gap-bridging nerve tissue. Although macroscopic tissue regeneration was not further improved after combining ESTIM with FGF-2(21/23-kD) gene therapy, the latter resulted in a high rate of regenerated nerves that functionally reconnected to the target muscle. Based on our results, brief ESTIM shows high potential to accelerate axonal as well as functional (motor and sensory) outcomes in the clinical setting of peripheral nerve gap reconstruction in human patients.


Assuntos
Axônios/fisiologia , Estimulação Elétrica , Regeneração Nervosa/fisiologia , Recuperação de Função Fisiológica/fisiologia , Nervo Isquiático/fisiologia , Animais , Feminino , Atividade Motora/fisiologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Schwann/fisiologia , Células de Schwann/transplante , Nervo Isquiático/transplante , Estatísticas não Paramétricas , Transplante Autólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA