Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 72018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30412052

RESUMO

Insulin gene mutations are a leading cause of neonatal diabetes. They can lead to proinsulin misfolding and its retention in endoplasmic reticulum (ER). This results in increased ER-stress suggested to trigger beta-cell apoptosis. In humans, the mechanisms underlying beta-cell failure remain unclear. Here we show that misfolded proinsulin impairs developing beta-cell proliferation without increasing apoptosis. We generated induced pluripotent stem cells (iPSCs) from people carrying insulin (INS) mutations, engineered isogenic CRISPR-Cas9 mutation-corrected lines and differentiated them to beta-like cells. Single-cell RNA-sequencing analysis showed increased ER-stress and reduced proliferation in INS-mutant beta-like cells compared with corrected controls. Upon transplantation into mice, INS-mutant grafts presented reduced insulin secretion and aggravated ER-stress. Cell size, mTORC1 signaling, and respiratory chain subunits expression were all reduced in INS-mutant beta-like cells, yet apoptosis was not increased at any stage. Our results demonstrate that neonatal diabetes-associated INS-mutations lead to defective beta-cell mass expansion, contributing to diabetes development.


Assuntos
Diabetes Mellitus/genética , Estresse do Retículo Endoplasmático/genética , Células-Tronco Pluripotentes Induzidas/química , Proinsulina/genética , Animais , Apoptose/genética , Sistemas CRISPR-Cas/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Diabetes Mellitus/patologia , Retículo Endoplasmático/genética , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Recém-Nascido , Células Secretoras de Insulina/química , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Mutação , Proinsulina/química , Dobramento de Proteína , Análise de Sequência de RNA , Transdução de Sinais , Análise de Célula Única
2.
Sci Adv ; 4(5): eaap8957, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29806020

RESUMO

Stroke is the most common cause of adult disability in developed countries, largely because spontaneous recovery is often incomplete, and no pharmacological means to hasten the recovery exist. It was recently shown that mesencephalic astrocyte-derived neurotrophic factor (MANF) induces alternative or M2 activation of immune cells after retinal damage in both fruit fly and mouse and mediates retinal repair. Therefore, we set out to study whether poststroke MANF administration would enhance brain tissue repair and affect behavioral recovery of rats after cerebral ischemic injury. We used the distal middle cerebral artery occlusion (dMCAo) model of ischemia-reperfusion injury and administered MANF either as a recombinant protein or via adeno-associated viral (AAV) vector. We discovered that, when MANF was administered to the peri-infarct region 2 or 3 days after stroke, it promoted functional recovery of the animals without affecting the lesion volume. Further, AAV7-MANF treatment transiently increased the number of phagocytic macrophages in the subcortical peri-infarct regions. In addition, the analysis of knockout mice revealed the neuroprotective effects of endogenous MANF against ischemic injury, although endogenous MANF had no effect on immune cell-related gene expression. The beneficial effect of MANF treatment on the reversal of stroke-induced behavioral deficits implies that MANF-based therapies could be used for the repair of brain tissue after stroke.


Assuntos
Astrócitos/metabolismo , Fatores de Crescimento Neural/genética , Reabilitação do Acidente Vascular Cerebral , Acidente Vascular Cerebral/metabolismo , Animais , Comportamento Animal , Isquemia Encefálica/complicações , Dependovirus/genética , Modelos Animais de Doenças , Expressão Gênica , Vetores Genéticos/genética , Humanos , Imageamento por Ressonância Magnética , Masculino , Fatores de Crescimento Neural/metabolismo , Ratos , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/etiologia , Transdução Genética , Transgenes
3.
Behav Brain Res ; 291: 1-11, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25975173

RESUMO

Cerebral dopamine neurotrophic factor (CDNF) protects and repairs dopamine neurons in animal models of Parkinson's disease, which motivated us to investigate its therapeutic effect in an animal model of Alzheimer's disease (AD). We employed an established APP/PS1 mouse model of AD and gave intrahippocampal injections of CDNF protein or CDNF transgene in an AAV2 viral vector to 1-year-old animals. We performed a behavioral test battery 2 weeks after the injections and collected tissue samples after the 3-week test period. Intrahippocampal CDNF-therapy improved long-term memory in both APP/PS1 mice and wild-type controls, but did not affect spontaneous exploration, object neophobia or early stages of spatial learning. The memory improvement was not associated with decreased brain amyloid load or enhanced hippocampal neurogenesis. Intracranial CDNF treatment has beneficial effects on long-term memory and is well tolerated. The CDNF molecular mechanisms of action on memory await further studies.


Assuntos
Doença de Alzheimer/metabolismo , Hipocampo/metabolismo , Memória de Longo Prazo/fisiologia , Fatores de Crescimento Neural/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Doença de Alzheimer/terapia , Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Terapia Genética/métodos , Vetores Genéticos , Hipocampo/patologia , Humanos , Consolidação da Memória/fisiologia , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores de Crescimento Neural/genética , Neurogênese/fisiologia , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
Dev Neurobiol ; 70(5): 360-71, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20186704

RESUMO

Current therapeutic interventions for neurodegenerative diseases alleviate only disease symptoms, while treatments that could stop or reverse actual degenerative processes are not available. Parkinson's disease (PD) is a movement disorder with characteristic degeneration of dopaminergic neurons in the midbrain. Few neurotrophic factors (NTFs) that promote survival, maintenance, and differentiation of affected brain neurons are considered as potential therapeutic agents for the treatment of neurodegenerative diseases. Thus, it is important to search and study new NTFs that could also be used in therapy. In this review, we discuss novel evolutionary conserved family of NTFs consisting of two members in the vertebrates, cerebral dopamine neurotrophic factor (CDNF) and mesencephalic astrocyte-derived neurotrophic factor (MANF). Invertebrates, including Drosophila and Caenorhabditis have a single protein homologous to vertebrate CDNF/MANF. Characteristic feature of these proteins is eight structurally conserved cysteine residues, which determine the protein fold. The crystal structure analysis revealed that CDNF and MANF consist of two domains; an amino-terminal saposin-like domain that may interact with lipids or membranes, and a presumably unfolded carboxy-terminal domain that may protect cells against endoplasmic reticulum stress. CDNF and MANF protect midbrain dopaminergic neurons and restore motor function in 6-hydroxydopamine rat model of PD in vivo. In line, Drosophila MANF is needed for the maintenance of dopaminergic neurites and dopamine levels in the fly, suggesting that the function of CDNF/MANF proteins is evolutionary conserved. Future studies will reveal the receptors and mode of action of these novel factors, which are potential therapeutic proteins for the treatment of PD.


Assuntos
Fatores de Crescimento Neural/metabolismo , Animais , Dopamina/metabolismo , Retículo Endoplasmático/fisiologia , Humanos , Modelos Neurológicos , Fatores de Crescimento Neural/química , Fatores de Crescimento Neural/genética , Neurônios/fisiologia , Estresse Fisiológico/fisiologia
5.
Cell Mol Neurobiol ; 30(3): 361-8, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19777340

RESUMO

Available treatment for Parkinson's disease (PD) is mainly symptomatic instead of halting or reversing degenerative processes affecting the disease. Research on the molecular pathogenesis of PD has suggested reduced trophic support as a possible cause or mediator of neurodegeneration. In animal models of the disease, neurotrophic factors prevent neurodegeneration and induce behavioral recovery. Some anti-Parkinsonian drugs show neuroprotective activity, but it is not known whether the drug-induced neuroprotection is mediated by neurotrophic factors. In this study, we have investigated the influence of two neuroprotective anti-Parkinsonian drugs, the monoamine oxidase B inhibitor selegiline and the adenosine A(2A) antagonist SCH 58261, on the levels of brain-derived neurotrophic factor (BDNF) and cerebral dopamine neurotrophic factor (CDNF) in the mouse brain. Protein levels of BDNF and CDNF were quantified by western blot after 2 weeks of treatment with either of the drugs or placebo. CDNF levels were not significantly influenced by selegiline or SCH 58261 in any brain area studied. Selegiline treatment significantly increased BDNF levels in the anterior cingulate cortex (1.55 +/- 0.22, P < 0.05, Student's t-test). In the striatum, selegiline increased BDNF content by 32%, but this change did not reach statistical significance (1.32 +/- 0.15, P < 0.13, Student's t-test). Our data suggest that neurotrophic factors, particularly BDNF may play a role in the neuroprotective effects of selegiline, but do not support the hypothesis that anti-Parkinsonian drugs would work by increasing the levels of CDNF in brain.


Assuntos
Antiparkinsonianos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Fatores de Crescimento Neural/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/tratamento farmacológico , Antagonistas do Receptor A2 de Adenosina , Animais , Western Blotting , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Citoproteção/efeitos dos fármacos , Citoproteção/fisiologia , Giro do Cíngulo/efeitos dos fármacos , Giro do Cíngulo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibidores da Monoaminoxidase/farmacologia , Fatores de Crescimento Neural/metabolismo , Pirimidinas/farmacologia , Receptor A2A de Adenosina/metabolismo , Selegilina/farmacologia , Triazóis/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
6.
Ann Neurol ; 62(3): 278-87, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17823937

RESUMO

OBJECTIVE: We studied the prevalence, segregation, and phenotype of the mitochondrial DNA 3243A>G mutation in children in a defined population in Northern Ostrobothnia, Finland. METHODS: Children with diagnoses commonly associated with mitochondrial diseases were ascertained. Blood DNA from 522 selected children was analyzed for 3243A>G. Children with the mutation were clinically examined. Information on health history before the age of 18 years was collected from previously identified adult patients with 3243A>G. Mutation segregation analysis in buccal epithelial cells was performed in mothers with 3243A>G and their children whose samples were analyzed anonymously. RESULTS: Eighteen children were found to harbor 3243A>G in a population of 97,609. A minimum estimate for the prevalence of 3243A>G was 18.4 in 100,000 (95% confidence interval, 10.9-29.1/100,000). Information on health in childhood was obtained from 37 adult patients with 3243A>G. The first clinical manifestations appearing in childhood were sensorineural hearing impairment, short stature or delayed maturation, migraine, learning difficulties, and exercise intolerance. Mutation analysis from 13 mothers with 3243A>G and their 41 children gave a segregation rate of 0.80. The mothers with heteroplasmy greater than 50% tended to have offspring with lower or equal heteroplasmy, whereas the opposite was true for mothers with heteroplasmy less than or equal to 50% (p = 0.0016). INTERPRETATION: The prevalence of 3243A>G is relatively high in the pediatric population, but the morbidity in children is relatively low. The random genetic drift model may be inappropriate for the transmission of the 3243A>G mutation.


Assuntos
DNA Mitocondrial/genética , Doenças Mitocondriais/genética , Mutação/genética , Adulto , Estatura/genética , Criança , Segregação de Cromossomos/genética , Estudos de Coortes , DNA Mitocondrial/sangue , Células Epiteliais , Tolerância ao Exercício/genética , Feminino , Finlândia/epidemiologia , Frequência do Gene , Deriva Genética , Crescimento/genética , Crescimento/fisiologia , Haplótipos , Perda Auditiva Neurossensorial/genética , Humanos , Deficiências da Aprendizagem/genética , Síndrome MELAS/genética , Masculino , Transtornos de Enxaqueca/genética , Mucosa Bucal/citologia , Mutação/fisiologia , Fenótipo
7.
Nature ; 448(7149): 73-7, 2007 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-17611540

RESUMO

In Parkinson's disease, brain dopamine neurons degenerate most prominently in the substantia nigra. Neurotrophic factors promote survival, differentiation and maintenance of neurons in developing and adult vertebrate nervous system. The most potent neurotrophic factor for dopamine neurons described so far is the glial-cell-line-derived neurotrophic factor (GDNF). Here we have identified a conserved dopamine neurotrophic factor (CDNF) as a trophic factor for dopamine neurons. CDNF, together with its previously described vertebrate and invertebrate homologue the mesencephalic-astrocyte-derived neurotrophic factor, is a secreted protein with eight conserved cysteine residues, predicting a unique protein fold and defining a new, evolutionarily conserved protein family. CDNF (Armetl1) is expressed in several tissues of mouse and human, including the mouse embryonic and postnatal brain. In vivo, CDNF prevented the 6-hydroxydopamine (6-OHDA)-induced degeneration of dopaminergic neurons in a rat experimental model of Parkinson's disease. A single injection of CDNF before 6-OHDA delivery into the striatum significantly reduced amphetamine-induced ipsilateral turning behaviour and almost completely rescued dopaminergic tyrosine-hydroxylase-positive cells in the substantia nigra. When administered four weeks after 6-OHDA, intrastriatal injection of CDNF was able to restore the dopaminergic function and prevent the degeneration of dopaminergic neurons in substantia nigra. Thus, CDNF was at least as efficient as GDNF in both experimental settings. Our results suggest that CDNF might be beneficial for the treatment of Parkinson's disease.


Assuntos
Fatores de Crescimento Neural/fisiologia , Neurônios/fisiologia , Sequência de Aminoácidos , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Clonagem Molecular , Sequência Conservada , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Humanos , Hibridização In Situ , Masculino , Camundongos , Dados de Sequência Molecular , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Oxidopamina , Doença de Parkinson/tratamento farmacológico , Processamento de Proteína Pós-Traducional , RNA Mensageiro , Ratos , Ratos Wistar , Substância Negra/metabolismo
8.
FEBS Lett ; 569(1-3): 267-71, 2004 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-15225646

RESUMO

Previously, it was shown that the recruitment of RET into lipid rafts by glial cell line-derived neurotrophic factor (GDNF)/GFRalpha1 is crucial for efficient signal transduction. Here, we show that the mouse GFRalpha4 is a functional, N-glycosylated, glycosylphosphatidylinositol (GPI)-anchored protein, which mediates persephin (PSPN)-induced phosphorylation of RET, but has an almost undetectable capacity to recruit RET into the 0.1% Triton X-100 insoluble membrane fraction. In spite of this, PSPN/mGFRalpha4 promotes neurite outgrowth in PC6-3 cells and survival of cerebellar granule neurons. As we show that also human PSPN/GFRalpha4 is unable to recruit RET into lipid rafts, we propose that the mammalian GFRalpha4 in this respect differs from GFRalpha1.


Assuntos
Glicoproteínas de Membrana/metabolismo , Microdomínios da Membrana/metabolismo , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuritos/fisiologia , Neurônios/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Animais , Linhagem Celular , Membrana Celular/fisiologia , Clonagem Molecular , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Neurônios/citologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ret , Ratos , Receptores Proteína Tirosina Quinases/genética , Receptores de Fator de Crescimento Neural/genética , Proteínas Recombinantes/metabolismo , Transfecção
9.
J Gen Virol ; 83(Pt 5): 1211-1221, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11961277

RESUMO

Processing of the polyprotein encoded by Potato virus A (PVA; genus Potyvirus) was studied using expression of the complete PVA polyprotein or its mutants from recombinant baculoviruses in insect cells. The time-course of polyprotein processing by the main viral proteinase (NIaPro) was examined with the pulse-chase method. The sites at the P3/6K1, CI-6K2 and VPg/NIaPro junctions were processed slowly, in contrast to other proteolytic cleavage sites which were processed at a high rate. The CI-6K2 polyprotein was observed in the baculovirus system and in infected plant cells. In both cell types the majority of CI-6K2 was found in the membrane fraction, in contrast to fully processed CI. Deletion of the genomic region encoding the 6K1 protein prevented proper proteolytic separation of P3 from CI, but did not affect processing of VPg, NIaPro, NIb or CP from the polyprotein. The 6K2-encoding sequence could be removed without any detectable effect on polyprotein processing. However, deletion of either the 6K1 or 6K2 protein-encoding regions rendered PVA non-infectious. Mutations at the 6K2/VPg cleavage site reduced virus infectivity in plants, but had a less pronounced, albeit detectable, effect on proteolytic processing in the baculovirus system. The results of this study indicate that NIaPro catalyses proteolytic cleavages preferentially in cis, and that the 6K1/CI and NIb/CP sites can also be processed in trans. Both 6K peptides are indispensable for virus replication, and proteolytic separation of the 6K2 protein from the adjacent proteins by NIaPro is important for the rate of virus replication and movement.


Assuntos
Endopeptidases/fisiologia , Plantas/virologia , Poliproteínas/metabolismo , Potyvirus/metabolismo , Proteínas Virais/metabolismo , Animais , Baculoviridae/metabolismo , Células Cultivadas , Spodoptera , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA