Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Colloid Interface Sci ; 627: 283-298, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35853406

RESUMO

HYPOTHESIS: The positive charge on liposome surface is known to promote the crossing of the Blood brain barrier (BBB). However, when diastereomeric cationic gemini amphiphiles are among lipid membrane components, also the stereochemistry may affect the permeability of the vesicle across the BBB. EXPERIMENTS: Liposomes featuring cationic diasteromeric gemini amphiphiles were formulated, characterized, and their interaction with cell culture models of BBB investigated. FINDINGS: Liposomes featuring the gemini amphiphiles were internalized in a monolayer of brain microvascular endothelial cells derived from human induced pluripotent stem cells (hiPSC) through an energy dependent transport, internalization involving both clathrin- and caveolae-mediated endocytosis. On the same formulations, the permeability was also evaluated across a human derived in vitro BBB transport model. The permeability of liposomes featuring the gemini amphiphiles was significantly higher compared to that of neutral liposomes (DPPC/Cholesterol), that were not able to cross BBB. Most importantly, the permeability was influenced by the stereochemistry of the gemini and pegylation of these formulations did not result in a drastic reduction of the crossing ability. The in vitro iPSC-derived BBB models used in this work represent an important advancement in the drug discovery research of novel brain delivery strategies and therapeutics for central nervous system diseases.


Assuntos
Células-Tronco Pluripotentes Induzidas , Lipossomos , Transporte Biológico , Barreira Hematoencefálica , Cátions , Colesterol , Clatrina , Células Endoteliais , Humanos , Lipossomos/química
2.
Int J Mol Sci ; 23(14)2022 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-35887162

RESUMO

While blood-brain barrier (BBB) dysfunction has been described in neurological disorders, including Huntington's disease (HD), it is not known if endothelial cells themselves are functionally compromised when promoting BBB dysfunction. Furthermore, the underlying mechanisms of BBB dysfunction remain elusive given the limitations with mouse models and post mortem tissue to identify primary deficits. We established models of BBB and undertook a transcriptome and functional analysis of human induced pluripotent stem cell (iPSC)-derived brain-like microvascular endothelial cells (iBMEC) from HD patients or unaffected controls. We demonstrated that HD-iBMECs have abnormalities in barrier properties, as well as in specific BBB functions such as receptor-mediated transcytosis.


Assuntos
Doença de Huntington , Células-Tronco Pluripotentes Induzidas , Animais , Barreira Hematoencefálica/fisiologia , Diferenciação Celular , Células Endoteliais/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Camundongos
3.
Cells ; 9(4)2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32316221

RESUMO

The blood-brain barrier (BBB) is responsible for the homeostasis between the cerebral vasculature and the brain and it has a key role in regulating the influx and efflux of substances, in healthy and diseased states. Stem cell technology offers the opportunity to use human brain-specific cells to establish in vitro BBB models. Here, we describe the establishment of a human BBB model in a two-dimensional monolayer culture, derived from human induced pluripotent stem cells (hiPSCs). This model was characterized by a transendothelial electrical resistance (TEER) higher than 2000 Ω∙cm2 and associated with negligible paracellular transport. The hiPSC-derived BBB model maintained the functionality of major endothelial transporter proteins and receptors. Some proprietary molecules from our central nervous system (CNS) programs were evaluated revealing comparable permeability in the human model and in the model from primary porcine brain endothelial cells (PBECs).


Assuntos
Transporte Biológico/efeitos dos fármacos , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Astrócitos/metabolismo , Transporte Biológico/fisiologia , Encéfalo/citologia , Diferenciação Celular/fisiologia , Células Cultivadas , Sistema Nervoso Central/química , Sistema Nervoso Central/metabolismo , Criopreservação/métodos , Humanos , Imuno-Histoquímica , Permeabilidade , Suínos
4.
Mol Pharm ; 16(5): 2069-2082, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30916978

RESUMO

Huntington's disease (HD) is a neurodegenerative disease caused by polyglutamine expansion in the huntingtin protein. For drug candidates targeting HD, the ability to cross the blood-brain barrier (BBB) and reach the site of action in the central nervous system (CNS) is crucial for achieving pharmacological activity. To assess the permeability of selected compounds across the BBB, we utilized a two-dimensional model composed of primary porcine brain endothelial cells and rat astrocytes. Our objective was to use this in vitro model to rank and prioritize compounds for in vivo pharmacokinetic and brain penetration studies. The model was first characterized using a set of validation markers chosen based on their functional importance at the BBB. It was shown to fulfill the major BBB characteristics, including functional tight junctions, high transendothelial electrical resistance, expression, and activity of influx and efflux transporters. The in vitro permeability of 54 structurally diverse known compounds was determined and shown to have a good correlation with the in situ brain perfusion data in rodents. We used this model to investigate the BBB permeability of a series of new HD compounds from different chemical classes, and we found a good correlation with in vivo brain permeation, demonstrating the usefulness of the in vitro model for optimizing CNS drug properties and for guiding the selection of lead compounds in a drug discovery setting.


Assuntos
Barreira Hematoencefálica/metabolismo , Fármacos do Sistema Nervoso Central/uso terapêutico , Descoberta de Drogas/métodos , Doença de Huntington/tratamento farmacológico , Modelos Biológicos , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Astrócitos/metabolismo , Permeabilidade Capilar/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Técnicas de Cocultura , Impedância Elétrica , Células Endoteliais/metabolismo , Permeabilidade , Ratos , Ratos Sprague-Dawley , Proteínas Carreadoras de Solutos/metabolismo , Suínos , Junções Íntimas/metabolismo
5.
Autophagy ; 13(4): 654-669, 2017 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-28368777

RESUMO

Mitophagy is a highly specialized process to remove dysfunctional or superfluous mitochondria through the macroautophagy/autophagy pathway, aimed at protecting cells from the damage of disordered mitochondrial metabolism and apoptosis induction. PINK1, a neuroprotective protein mutated in autosomal recessive Parkinson disease, has been implicated in the activation of mitophagy by selectively accumulating on depolarized mitochondria, and promoting PARK2/Parkin translocation to them. While these steps have been characterized in depth, less is known about the process and site of autophagosome formation upon mitophagic stimuli. A previous study reported that, in starvation-induced autophagy, the proautophagic protein BECN1/Beclin1 (which we previously showed to interact with PINK1) relocalizes at specific regions of contact between the endoplasmic reticulum (ER) and mitochondria called mitochondria-associated membranes (MAM), from which the autophagosome originates. Here we show that, following mitophagic stimuli, autophagosomes also form at MAM; moreover, endogenous PINK1 and BECN1 were both found to relocalize at MAM, where they promoted the enhancement of ER-mitochondria contact sites and the formation of omegasomes, that represent autophagosome precursors. PARK2 was also enhanced at MAM following mitophagy induction. However, PINK1 silencing impaired BECN1 enrichment at MAM independently of PARK2, suggesting a novel role for PINK1 in regulating mitophagy. MAM have been recently implicated in many key cellular events. In this light, the observed prevalent localization of PINK1 at MAM may well explain other neuroprotective activities of this protein, such as modulation of mitochondrial calcium levels, mitochondrial dynamics, and apoptosis.


Assuntos
Autofagossomos/metabolismo , Proteína Beclina-1/metabolismo , Retículo Endoplasmático/metabolismo , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Mitofagia , Proteínas Quinases/metabolismo , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Compartimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação para Baixo , Humanos
6.
Stem Cells ; 32(9): 2516-28, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24806549

RESUMO

Adult neurogenesis is a multistep process regulated by several extrinsic factors, including neurotrophins. Among them, little is known about the role of nerve growth factor (NGF) in the neurogenic niches of the mouse. Here we analyzed the biology of adult neural stem cells (NSCs) from the subventricular zone (SVZ) of AD11 anti-NGF transgenic mice, in which the expression of the recombinant antibody aD11 leads to a chronic postnatal neutralization of endogenous NGF. We showed that AD11-NSCs proliferate 10-fold less, with respect to their control counterparts, and display a significant impairment in their ability to differentiate into ß-tubulin positive neurons. We found a considerable reduction in the number of SVZ progenitors and neuroblasts also in vivo, which correlates with a lower number of newborn neurons in the olfactory bulbs of AD11 mice and a severe deficit in the ability of these mice to discriminate between different odors. We also demonstrated that, in AD11 mice, the morphology of both SVZ-resident and neurosphere-derived astrocytes is significantly altered. We were able to reproduce the AD11 phenotype in vitro, by acutely treating wild type NSCs with the anti-NGF antibody, further demonstrating that both the proliferation and the differentiation defects are due to the NGF deprivation. Consistently, the proliferative impairment of AD11 progenitors, as well as the atrophic morphology of AD11 astrocytes, can be partly rescued in vitro and in vivo by exogenous NGF addition. Altogether, our results demonstrate a causal link between NGF signaling and proper proliferation and differentiation of neural stem cells from the SVZ.


Assuntos
Ventrículos Laterais/metabolismo , Fator de Crescimento Neural/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Ventrículos Laterais/citologia , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Neurônios/citologia , Transdução de Sinais
7.
Biochem Biophys Res Commun ; 431(3): 579-85, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23313508

RESUMO

Sortilin is a member of the family of vacuolar protein sorting 10 protein domain receptors which has emerged as a co-receptor in cell death and neurodegeneration processes mediated by proneurotrophins. Here we tested the possibility that sortilin deficiency interferes with behavioral and neuropathological endpoints in a chronic Nerve Growth factor (NGF)-deprivation model of Alzheimer's disease (AD), the AD10 anti-NGF mouse. AD10 mice show cholinergic deficit, increased APP processing and tau hyper-phosphorylation, resulting in behavioral deficits in learning and memory paradigms assessed by novel object recognition and Morris water maze tests. Sort1(-/-) mice were crossed with AD10 anti-NGF mice and the neurodegenerative phenotype was studied. We found that the loss of sortilin partially protected AD10 anti-NGF mice from neurodegeneration. A protective effect was observed on non-spatial memory as assessed by novel object recognition, and histopathologically at the level of Aß and BFCNs, while the phosphotau increase was unaltered by knocking out sortilin. We suggest that sortilin might be involved in different aspects of neurodegeneration in a complex way, supporting the view that sortilin functions in the CNS are broader than being a co-receptor in proneurotrophin and neurotrophin signaling.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Doença de Alzheimer/metabolismo , Fator de Crescimento Neural/deficiência , Proteínas Adaptadoras de Transporte Vesicular/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Colina/metabolismo , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/patologia , Modelos Animais de Doenças , Transtornos da Memória/genética , Camundongos , Camundongos Mutantes , Neurite (Inflamação)/genética , Fosforilação , Modificação Traducional de Proteínas , Receptor trkA/biossíntese , Transdução de Sinais , Proteínas tau/metabolismo
8.
J Alzheimers Dis ; 33(2): 357-71, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22986780

RESUMO

Sortilin-related receptor with A-type repeats (SorLA, also known as LR11) has been implicated in Alzheimer's disease (AD). Thus, genetic studies associated SorLA gene variants with the risk of sporadic AD. Also, in vitro and in vivo studies showed that SorLA impairs processing of the amyloid-ß protein precursor (AßPP) to amyloid-ß. In particular, it has been found that loss of SorLA accelerates senile plaque deposition in mouse models overexpressing mutant forms of human AßPP and presenilin 1. Here we tested the possibility that SorLA deficiency also interferes with behavioral and neuropathological endpoints in an alternative murine AD model, the AD10 anti-nerve growth factor (NGF) mouse, in which amyloid-ß accumulation derives from the altered processing of endogenous AßPP. In addition to alterations in AßPP processing, AD10 mice also show cholinergic deficit and tau hyperphosphorylation resulting in behavioral deficits in learning and memory paradigms. We found that the loss of SorLA not only exacerbates early amyloid pathology but, at the same time, protects from cholinergic deficit and from early phospho-tau mislocalization. The results show that in the AD10 anti-NGF mouse model the AßPP processing-related aspects of neurodegeneration can be dissociated from those related to tau posttranslational processing and to cholinergic phenotypic maintenance by modulation of SorLA expression. We suggest that SorLA regulates different aspects of neurodegeneration in a complex way, supporting the hypothesis that SorLA expression might be critical not only for amyloid-related pathology but also for other cellular processes altered in AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Amiloidose/genética , Neurônios Colinérgicos/metabolismo , Proteínas de Membrana Transportadoras/genética , Receptores de LDL/genética , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Amiloide/metabolismo , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Córtex Cerebral/patologia , Córtex Cerebral/fisiologia , Neurônios Colinérgicos/patologia , Modelos Animais de Doenças , Feminino , Masculino , Aprendizagem em Labirinto/fisiologia , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/metabolismo , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Fosforilação/fisiologia , Receptor trkA/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/metabolismo
9.
PLoS One ; 7(5): e37555, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22666365

RESUMO

Nerve Growth Factor (NGF) is being considered as a therapeutic candidate for Alzheimer's disease (AD) treatment but the clinical application is hindered by its potent pro-nociceptive activity. Thus, to reduce systemic exposure that would induce pain, in recent clinical studies NGF was administered through an invasive intracerebral gene-therapy approach. Our group demonstrated the feasibility of a non-invasive intranasal delivery of NGF in a mouse model of neurodegeneration. NGF therapeutic window could be further increased if its nociceptive effects could be avoided altogether. In this study we exploit forms of NGF, mutated at residue R100, inspired by the human genetic disease HSAN V (Hereditary Sensory Autonomic Neuropathy Type V), which would allow increasing the dose of NGF without triggering pain. We show that "painless" hNGF displays full neurotrophic and anti-amyloidogenic activities in neuronal cultures, and a reduced nociceptive activity in vivo. When administered intranasally to APPxPS1 mice ( n = 8), hNGFP61S/R100E prevents the progress of neurodegeneration and of behavioral deficits. These results demonstrate the in vivo neuroprotective and anti-amyloidogenic properties of hNGFR100 mutants and provide a rational basis for the development of "painless" hNGF variants as a new generation of therapeutics for neurodegenerative diseases.


Assuntos
Amiloide/metabolismo , Progressão da Doença , Transtornos da Memória/prevenção & controle , Fator de Crescimento Neural/administração & dosagem , Fator de Crescimento Neural/farmacologia , Doenças Neurodegenerativas/tratamento farmacológico , Nociceptividade/efeitos dos fármacos , Administração Intranasal , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Aprendizagem/efeitos dos fármacos , Aprendizagem/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Mutação , Fator de Crescimento Neural/efeitos adversos , Fator de Crescimento Neural/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/fisiopatologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/efeitos adversos , Fármacos Neuroprotetores/farmacologia , Fosfolipase C gama/metabolismo , Multimerização Proteica , Estrutura Quaternária de Proteína , Receptor trkA/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinaptofisina/metabolismo
10.
Proc Natl Acad Sci U S A ; 109(6): 2009-14, 2012 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22308471

RESUMO

Nerve growth factor (NGF) was discovered because of its neurotrophic actions on sympathetic and sensory neurons in the developing chicken embryo. NGF was subsequently found to influence and regulate the function of many neuronal and non neuronal cells in adult organisms. Little is known, however, about the possible actions of NGF during early embryonic stages. However, mRNAs encoding for NGF and its receptors TrkA and p75(NTR) are expressed at very early stages of avian embryo development, before the nervous system is formed. The question, therefore, arises as to what might be the functions of NGF in early chicken embryo development, before its well-established actions on the developing sympathetic and sensory neurons. To investigate possible roles of NGF in the earliest stages of development, stage HH 11-12 chicken embryos were injected with an anti-NGF antibody (mAb αD11) that binds mature NGF with high affinity. Treatment with anti-NGF, but not with a control antibody, led to a dose-dependent inversion of the direction of axial rotation. This effect of altered rotation after anti NGF injection was associated with an increased cell death in somites. Concurrently, a microarray mRNA expression analysis revealed that NGF neutralization affects the expression of genes linked to the regulation of development or cell proliferation. These results reveal a role for NGF in early chicken embryo development and, in particular, in the regulation of somite survival and axial rotation, a crucial developmental process linked to left-right asymmetry specification.


Assuntos
Padronização Corporal , Fator de Crescimento Neural/metabolismo , Rotação , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/genética , Proliferação de Células/efeitos dos fármacos , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Imuno-Histoquímica , Fator de Crescimento Neural/genética , Testes de Neutralização , Análise de Sequência com Séries de Oligonucleotídeos , Receptor trkA/genética , Receptor trkA/metabolismo , Somitos/efeitos dos fármacos , Somitos/embriologia
11.
Proc Natl Acad Sci U S A ; 107(27): 12299-304, 2010 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-20566851

RESUMO

NGF, the principal neurotrophic factor for basal forebrain cholinergic neurons (BFCNs), has been correlated to Alzheimer's disease (AD) because of the selective vulnerability of BFCNs in AD. These correlative links do not substantiate a comprehensive cause-effect mechanism connecting NGF deficit to overall AD neurodegeneration. A demonstration that neutralizing NGF activity could have consequences beyond a direct interference with the cholinergic system came from studies in the AD11 mouse model, in which the expression of a highly specific anti-NGF antibody determines a neurodegeneration that encompasses several features of human AD. Because the transgenic antibody binds to mature NGF much more strongly than to proNGF and prevents binding of mature NGF to the tropomyosin-related kinase A (TrkA) receptor and to p75 neurotrophin receptor (p75NTR), we postulated that neurodegeneration in AD11 mice is provoked by an imbalance of proNGF/NGF signaling and, consequently, of TrkA/p75NTR signaling. To test this hypothesis, in this study we characterize the phenotype of two lines of transgenic mice, one in which TrkA signaling is inhibited by neutralizing anti-TrkA antibodies and a second one in which anti-NGF mice were crossed to p75NTR(exonIII(-/-)) mice to abrogate p75NTR signaling. TrkA neutralization determines a strong cholinergic deficit and the appearance of beta-amyloid peptide (Abeta) but no tau-related pathology. In contrast, abrogating p75NTR signaling determines a full rescue of the cholinergic and Abeta phenotype of anti-NGF mice, but tau hyperphosphorylation is exacerbated. Thus, we demonstrate that inhibiting TrkA signaling activates Abeta accumulation and that different streams of AD neurodegeneration are related in complex ways to TrkA versus p75NTR signaling.


Assuntos
Doença de Alzheimer/metabolismo , Fator de Crescimento Neural/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Fatores Etários , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Fator de Crescimento Neural/deficiência , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/imunologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Ligação Proteica , Receptor de Fator de Crescimento Neural/genética , Receptor de Fator de Crescimento Neural/imunologia , Receptor trkA/genética , Transdução de Sinais , Proteínas tau/metabolismo
12.
J Alzheimers Dis ; 20(2): 527-46, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20182028

RESUMO

We previously showed that anti-nerve growth factor (NGF) antibodies expressed in transgenic mice (AD11) elicit a progressive neurodegeneration, comprising the triad of Alzheimer's disease (AD) hallmarks: cholinergic loss, tau hyperphosphorylation, and amyloid-beta peptide formation. However, since anti-NGF antibodies are produced both in the brain and in peripheral tissues of AD11 mice, the contribution of peripheral neutralization of NGF to the onset of brain neurodegeneration was still unexplored. To address this question, we characterized a line of transgenic mice (AD10) in which anti-NGF antibodies are obligatorily produced only in lymphocytes, being initially found in blood. In AD10 mice, peripheral NGF neutralization elicits shrinkage of superior cervical ganglia (immunosympathectomy) and, as a consequence of this, peripheral anti-NGF antibodies cross the blood brain barrier (BBB) and reach the brain, generating an NGF-dependent neurodegeneration, largely superimposable to that observed in AD11 mice. This demonstrates that peripherally originated anti-NGF antibodies can generate a neurodegeneration in the central nervous system of an animal model. Consistently, peripherally-delivered NGF is effective in preventing the onset of the central cholinergic deficit. These findings could have a direct relevance for some human sporadic AD cases, highlighting the role of the BBB disruption and suggesting a causally relevant role of circulating antibodies in AD pathology.


Assuntos
Anticorpos/uso terapêutico , Autoanticorpos/genética , Degeneração Neural/tratamento farmacológico , Degeneração Neural/imunologia , Fator de Crescimento Neural/imunologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Anticorpos/metabolismo , Autoanticorpos/imunologia , Barreira Hematoencefálica/fisiopatologia , Células COS , Chlorocebus aethiops , Colina O-Acetiltransferase/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Cadeias Leves de Imunoglobulina/genética , Cadeias Leves de Imunoglobulina/metabolismo , Transtornos da Memória/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Degeneração Neural/complicações , Degeneração Neural/patologia , Fator de Crescimento Neural/administração & dosagem , Testes Neuropsicológicos , Sistema Nervoso Parassimpático/fisiologia , Reconhecimento Psicológico/fisiologia , Gânglio Cervical Superior/efeitos dos fármacos , Gânglio Cervical Superior/patologia , Transfecção/métodos , Tirosina 3-Mono-Oxigenase/metabolismo
13.
Biochem Biophys Res Commun ; 391(1): 824-9, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19945432

RESUMO

Nerve Growth Factor (NGF) signalling is mediated by the TrkA and p75NTR receptors. Besides its neurotrophic and survival activities, NGF displays a potent pro-nociceptive activity. Recently, a missense point mutation was found in the NGFB gene (C661T, leading to the aminoacid substitution R100W) of individuals affected by a form of hereditary loss of pain perception (hereditary sensory and autonomic neuropathy type V, HSAN V). In order to gain insights into the functional consequences of the HSAN V NGF mutation, two sets of hNGFR100 mutants were expressed in Escherichia coli and purified, as mature NGF or proNGF, for in vitro receptor binding studies. Here, we show by Surface Plasmon Resonance analysis that the R100 mutation selectively disrupts binding of hNGF to p75NTR receptor, to an extent which depends on the substituting residue at position 100, while the affinity of hNGFR100 mutants for TrkA receptor is not affected. As for unprocessed hproNGF, the binding of the R100 variants to p75NTR receptor shows only a limited impairment, showing that the impact of the R100 mutation on p75NTR receptor binding is greater in the context of mature, processed hNGF. These results provide a basis for elucidating the mechanisms underlying the clinical manifestations of HSAN V patients, and provide a basis for the development of "painless" hNGF molecules with therapeutic potential.


Assuntos
Neuropatias Hereditárias Sensoriais e Autônomas/metabolismo , Fator de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Insensibilidade Congênita à Dor/metabolismo , Receptor trkA/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Neuropatias Hereditárias Sensoriais e Autônomas/genética , Humanos , Mutação , Fator de Crescimento Neural/química , Fator de Crescimento Neural/genética , Insensibilidade Congênita à Dor/genética , Conformação Proteica , Ressonância de Plasmônio de Superfície
14.
J Alzheimers Dis ; 16(2): 371-88, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19221427

RESUMO

Nerve growth factor (NGF) has a great potential for the treatment of Alzheimer's disease. However, the therapeutic administration of NGF represents a significant challenge, due to the difficulty to deliver relevant doses to the brain, in a safe and non-invasive way. We previously demonstrated the efficacy of a non-invasive delivery of NGF to the brain in animal models, by an intranasal route. Recently, topical eye application of NGF was proposed, as an option for the delivery of NGF to the brain. Here, we compare the efficacy of the two delivery routes of hNGF-61, a recombinant traceable form of human NGF, in the mouse neurodegeneration model AD11. The intranasal administration appeared to be significantly more effective than the ocular one, in rescuing the neurodegenerative phenotypic hallmarks in AD11 mice. The ocular administration of hNGF-61 showed a more limited efficacy, even at higher doses. Thus, NGF nasal drops represent a viable and effective option to successfully deliver therapeutic NGF to the brain in a non-invasive manner.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Anticorpos Monoclonais/administração & dosagem , Fator de Crescimento Neural/imunologia , Administração Intranasal , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Sequência de Aminoácidos , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/enzimologia , Colina O-Acetiltransferase/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Esquema de Medicação , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Camundongos , Camundongos Knockout , Microinjeções , Mutagênese , Fator de Crescimento Neural/deficiência , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/uso terapêutico , Reconhecimento Visual de Modelos/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA