Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Intervalo de año de publicación
1.
eNeuro ; 11(9)2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39293937

RESUMEN

Retinitis pigmentosa (RP) is a family of genetically heterogeneous diseases still without a cure. Despite the causative genetic mutation typically not expressed in cone photoreceptors, these cells inevitably degenerate following the primary death of rods, causing blindness. The reasons for the "bystander" degeneration of cones are presently unknown but decrement of survival factors, oxidative stress, and inflammation all play a role. Targeting these generalized biological processes represents a strategy to develop mutation-agnostic therapies for saving vision in large populations of RP individuals. A classical method to support neuronal survival is by employing neurotrophic factors, such as NGF. This study uses painless human NGF (hNGFp), a TrkA receptor-biased variant of the native molecule with lower affinity for nociceptors and limited activity as a pain inducer; the molecule has identical neurotrophic power of the native form but a reduced affinity for the p75NTR receptors, known to trigger apoptosis. hNGFp has a recognized activity on brain microglial cells, which are induced to a phenotype switch from a highly activated to a more homeostatic configuration. hNGFp was administered to RP-like mice in vivo with the aim of decreasing retinal inflammation and also providing retinal neuroprotection. However, the ability of this treatment to counteract the bystander degeneration of cones remained limited.


Asunto(s)
Factor de Crecimiento Nervioso , Retinitis Pigmentosa , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/genética , Animales , Factor de Crecimiento Nervioso/administración & dosificación , Factor de Crecimiento Nervioso/metabolismo , Humanos , Retina/metabolismo , Ratones Endogámicos C57BL , Ratones , Modelos Animales de Enfermedad , Receptor trkA/metabolismo , Masculino , Femenino , Microglía/metabolismo , Microglía/efectos de los fármacos
2.
Hum Mol Genet ; 32(8): 1380-1400, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-36537577

RESUMEN

A functional nerve growth factor NGF-Tropomyosin Receptor kinase A (TrkA) system is an essential requisite for the generation and maintenance of long-lasting thermal and mechanical hyperalgesia in adult mammals. Indeed, mutations in the gene encoding for TrkA are responsible for a rare condition, named Hereditary Sensory and Autonomic Neuropathy type IV (HSAN IV), characterized by the loss of response to noxious stimuli, anhidrosis and cognitive impairment. However, to date, there is no available mouse model to properly understand how the NGF-TrkA system can lead to pathological phenotypes that are distinctive of HSAN IV. Here, we report the generation of a knock-in mouse line carrying the HSAN IV TrkAR649W mutation. First, by in vitro biochemical and biophysical analyses, we show that the pathological R649W mutation leads to kinase-inactive TrkA also affecting its membrane dynamics and trafficking. In agreement with the HSAN IV human phenotype, TrkAR649W/m mice display a lower response to thermal and chemical noxious stimuli, correlating with reduced skin innervation, in addition to decreased sweating in comparison to TrkAh/m controls. Moreover, the R649W mutation decreases anxiety-like behavior and compromises cognitive abilities, by impairing spatial-working and social memory. Our results further uncover unexplored roles of TrkA in thermoregulation and sociability. In addition to accurately recapitulating the clinical manifestations of HSAN IV patients, our findings contribute to clarifying the involvement of the NGF-TrkA system in pain sensation.


Asunto(s)
Modelos Animales de Enfermedad , Neuropatías Hereditarias Sensoriales y Autónomas , Receptor trkA , Humanos , Animales , Ratones , Mutación , Receptor trkA/genética , Técnicas de Sustitución del Gen , Factor de Crecimiento Nervioso/metabolismo , Fosforilación , Genes Letales , Dolor/metabolismo , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Piel/metabolismo , Piel/patología , Sistema Nervioso Simpático/metabolismo , Hipohidrosis/metabolismo , Conducta Animal
3.
Pharmacol Res ; 139: 17-25, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30391352

RESUMEN

Nerve Growth Factor (NGF) is a therapeutic candidate for Alzheimer's disease, based on its well known actions on basal forebrain cholinergic neurons. However, because of its pro-nociceptive activity, in current clinical trials NGF has to be administered intraparenchymally into the brain by neurosurgery via cell or gene therapy approaches. To prevent the NGF pain-inducing collateral effects, thus avoiding the necessity for local brain injection, we developed painless NGF (hNGFp), based on the human genetic disease Hereditary Sensory and Autonomic Neuropathy type V (HSAN V). hNGFp has similar neurotrophic activity as wild type human NGF, but its pain sensitizing activity is tenfold lower. Pharmacologically, hNGFp is a biased receptor agonist of NGF TrkA receptor. The results of recent studies shed new light on the neuroprotective mechanism by hNGFp and are highly relevant for the planning of NGF-based clinical trials. The intraparenchymal delivery of hNGFp, as used in clinical trials, was simulated in the 5xFAD mouse model and found to be inefficacious in reducing Aß plaque load. On the contrary, the same dose of hNGFp administered intranasally, which was rather widely biodistributed in the brain and did not induce pain sensitization, blocked APP processing into amyloid and restored synaptic plasticity and memory in this aggressive neurodegeneration model. This potent and broad neuroprotection by hNGFp was found to be mediated by hNGFp actions on glial cells. hNGFp increases inflammatory proteins such as the soluble TNFα receptor II and the chemokine CXCL12. Independent work has shown that NGF has a potent anti-inflammatory action on microglia and steers them towards a neuroprotective phenotype. These studies demonstrate that microglia cells are a new target cell of NGF in the brain and have therapeutic significance: i) they establish that the neuroprotective actions of hNGFp relies on a widespread exposure of the brain, ii) they identify a new anti-neurodegenerative pathway, linking hNGFp to inflammatory chemokines and cytokines via microglia, a common target for new therapeutic opportunities for neurodegenerative diseases, iii) they extend the neuroprotective potential of hNGFp beyond its classical cholinergic target, thereby widening the range of neurological diseases for which this neurotrophic factor might be used therapeutically, iv) they help interpreting the results of current NGF clinical trials in AD and the design of future trials with this new potent therapeutic candidate.


Asunto(s)
Microglía/efectos de los fármacos , Factor de Crecimiento Nervioso/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Receptor trkA/agonistas , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Quimiocina CXCL12/metabolismo , Humanos , Microglía/metabolismo , Factor de Crecimiento Nervioso/efectos adversos , Factor de Crecimiento Nervioso/metabolismo , Neuroprotección , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/metabolismo , Dolor/inducido químicamente
4.
Neurobiol Dis ; 111: 36-47, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29246724

RESUMEN

α-synuclein (αS) is a small protein that self-aggregates into α-helical oligomer species and subsequently into larger insoluble amyloid fibrils that accumulate in intraneuronal inclusions during the development of Parkinson's disease. Toxicity of αS oligomers and fibrils has been long debated and more recent data are suggesting that both species can induce neurodegeneration. However while most of these data are based on differences in structure between oligomer and aggregates, often preassembled in vitro, the in vivo situation might be more complex and subcellular locations where αS species accumulate, rather than their conformation, might contribute to enhanced toxicity. In line with this observation, we have shown that αS oligomers and aggregates are associated with the endoplasmic reticulum/microsomes (ER/M) membrane in vivo and how accumulation of soluble αS oligomers at the ER/M level precedes neuronal degeneration in a mouse model of α-synucleinopathies. In this paper we took a further step, investigating the biochemical and functional features of αS species associated with the ER/M membrane. We found that by comparison with non-microsomal associated αS (P10), the ER/M-associated αS pool is a unique population of oligomers and aggregates with specific biochemical traits such as increased aggregation, N- and C-terminal truncations and phosphorylation at serine 129. Moreover, when administered to murine primary neurons, ER/M-associated αS species isolated from diseased A53T human αS transgenic mice induced neuronal changes in a time- and dose-dependent manner. In fact the addition of small amounts of ER/M-associated αS species from diseased mice to primary cultures induced the formation of beads-like structures or strings of fibrous αS aggregates along the neurites, occasionally covering the entire process or localizing at the soma level. By comparison treatment with P10 fractions from the same diseased mice resulted in the formation of scarce and small puncta only when administered at high amount. Moreover, increasing the amount of P100/M fractions obtained from diseased and, more surprisingly, from presymptomatic mice induced a significant level of neuronal death that was prevented when neurons were treated with ER/M fractions immunodepleted of αS high molecular weight (HMW) species. These data provide the first evidence of the existence of two different populations of αS HMW species in vivo, putting the spotlight on the association to ER/M membrane as a necessary step for the acquisition of αS toxic features.


Asunto(s)
Retículo Endoplásmico/metabolismo , Microsomas/metabolismo , Neuronas/metabolismo , Agregación Patológica de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Retículo Endoplásmico/patología , Humanos , Ratones Transgénicos , Peso Molecular , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Cultivo Primario de Células , Agregación Patológica de Proteínas/patología , alfa-Sinucleína/química , alfa-Sinucleína/genética
5.
PLoS One ; 10(9): e0136425, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26371475

RESUMEN

BACKGROUND: Nerve Growth Factor (NGF) holds a great therapeutic promise for Alzheimer's disease, diabetic neuropathies, ophthalmic diseases, dermatological ulcers. However, the necessity for systemic delivery has hampered the clinical applications of NGF due to its potent pro-nociceptive action. A "painless" human NGF (hNGF R100E) mutant has been engineered. It has equal neurotrophic potency to hNGF but a lower nociceptive activity. We previously described and characterized the neurotrophic and nociceptive properties also of the hNGF P61S and P61SR100E mutants, selectively detectable against wild type hNGF. However, the reduced pain-sensitizing potency of the "painless" hNGF mutants has not been quantified. OBJECTIVES AND RESULTS: Aiming at the therapeutic application of the "painless" hNGF mutants, we report on the comparative functional characterization of the precursor and mature forms of the mutants hNGF R100E and hNGF P61SR100E as therapeutic candidates, also in comparison to wild type hNGF and to hNGF P61S. The mutants were assessed by a number of biochemical, biophysical methods and assayed by cellular assays. Moreover, a highly sensitive ELISA for the detection of the P61S-tagged mutants in biological samples has been developed. Finally, we explored the pro-nociceptive effects elicited by hNGF mutants in vivo, demonstrating an expanded therapeutic window with a ten-fold increase in potency. CONCLUSIONS: This structure-activity relationship study has led to validate the concept of developing painless NGF as a therapeutic, targeting the NGF receptor system and supporting the choice of hNGF P61S R100E as the best candidate to advance in clinical development. Moreover, this study contributes to the identification of the molecular determinants modulating the properties of the hNGF "painless" mutants.


Asunto(s)
Mutación , Factor de Crecimiento Nervioso/efectos adversos , Factor de Crecimiento Nervioso/genética , Dolor/inducido químicamente , Ingeniería de Proteínas , Precursores de Proteínas/efectos adversos , Precursores de Proteínas/genética , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Escherichia coli/genética , Humanos , Cinética , Factor de Crecimiento Nervioso/metabolismo , Factor de Crecimiento Nervioso/uso terapéutico , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Precursores de Proteínas/metabolismo , Precursores de Proteínas/uso terapéutico , Estabilidad Proteica , Proteolisis , Ratas , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Temperatura
6.
Biochem Biophys Res Commun ; 431(3): 579-85, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23313508

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Enfermedad de Alzheimer/metabolismo , Factor de Crecimiento Nervioso/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Colina/metabolismo , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/patología , Modelos Animales de Enfermedad , Trastornos de la Memoria/genética , Ratones , Ratones Mutantes , Neuritis/genética , Fosforilación , Modificación Traduccional de las Proteínas , Receptor trkA/biosíntesis , Transducción de Señal , Proteínas tau/metabolismo
7.
Proc Natl Acad Sci U S A ; 110(5): 1935-40, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23319656

RESUMEN

Endocannabinoid, particularly 2-arachidonoyl glycerol (2-AG), signaling has recently emerged as a molecular determinant of neuronal migration and synapse formation during cortical development. However, the cell type specificity and molecular regulation of spatially and temporally confined morphogenic 2-AG signals remain unexplored. Here, we demonstrate that genetic and pharmacological manipulation of CB(1) cannabinoid receptors permanently alters cholinergic projection neuron identity and hippocampal innervation. We show that nerve growth factor (NGF), implicated in the morphogenesis and survival of cholinergic projection neurons, dose-dependently and coordinately regulates the molecular machinery for 2-AG signaling via tropomyosine kinase A receptors in vitro. In doing so, NGF limits the sorting of monoacylglycerol lipase (MGL), rate limiting 2-AG bioavailability, to proximal neurites, allowing cell-autonomous 2-AG signaling at CB(1) cannabinoid receptors to persist at atypical locations to induce superfluous neurite extension. We find that NGF controls MGL degradation in vitro and in vivo and identify the E3 ubiquitin ligase activity of breast cancer type 1 susceptibility protein (BRCA1) as a candidate facilitating MGL's elimination from motile neurite segments, including growth cones. BRCA1 inactivation by cisplatin or genetically can rescue and reposition MGL, arresting NGF-induced growth responses. These data indicate that NGF can orchestrate endocannabinoid signaling to promote cholinergic differentiation and implicate BRCA1 in determining neuronal morphology.


Asunto(s)
Endocannabinoides/metabolismo , Monoacilglicerol Lipasas/metabolismo , Factor de Crecimiento Nervioso/farmacología , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Ácidos Araquidónicos/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Glicéridos/metabolismo , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Monoacilglicerol Lipasas/genética , Neuronas/metabolismo , Células PC12 , Ratas , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
J Alzheimers Dis ; 33(2): 357-71, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22986780

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Amiloidosis/genética , Neuronas Colinérgicas/metabolismo , Proteínas de Transporte de Membrana/genética , Receptores de LDL/genética , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Amiloidosis/metabolismo , Amiloidosis/patología , Animales , Corteza Cerebral/patología , Corteza Cerebral/fisiología , Neuronas Colinérgicas/patología , Modelos Animales de Enfermedad , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/metabolismo , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Fosforilación/fisiología , Receptor trkA/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/metabolismo
9.
PLoS One ; 7(5): e37555, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22666365

RESUMEN

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.


Asunto(s)
Amiloide/metabolismo , Progresión de la Enfermedad , Trastornos de la Memoria/prevención & control , Factor de Crecimiento Nervioso/administración & dosificación , Factor de Crecimiento Nervioso/farmacología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Nocicepción/efectos de los fármacos , Administración Intranasal , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Ratones , Ratones Transgénicos , Modelos Moleculares , Mutación , Factor de Crecimiento Nervioso/efectos adversos , Factor de Crecimiento Nervioso/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/farmacología , Fosfolipasa C gamma/metabolismo , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Receptor trkA/metabolismo , Transducción de Señal/efectos de los fármacos , Sinaptofisina/metabolismo
10.
J Alzheimers Dis ; 31(1): 1-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22504318

RESUMEN

Several studies suggest that systemic infection occurring during aging and chronic neurodegenerative diseases can evoke an exaggerated immune response that contributes to the progression of neurodegeneration and cognitive decline. However, studies directly addressing the relationship between microbial environment and the onset of neurodegeneration in Alzheimer's disease animal models are lacking. Here we show that the onset of neurodegeneration that transgenic mice develop when raised in conventional husbandry slows down when raising anti-nerve growth factor transgenic mice in a murine pathogen free condition.


Asunto(s)
Factor de Crecimiento Nervioso/deficiencia , Enfermedades Neurodegenerativas/complicaciones , Enfermedades Neurodegenerativas/etiología , Animales , Infecciones por Caliciviridae/complicaciones , Colina O-Acetiltransferasa/metabolismo , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Interleucina-6/sangre , Lipopolisacáridos/toxicidad , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/sangre , Enfermedades Neurodegenerativas/patología , Norovirus/patogenicidad , Tricomoniasis/complicaciones , Proteínas tau/metabolismo
11.
Proc Natl Acad Sci U S A ; 109(6): 2009-14, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22308471

RESUMEN

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.


Asunto(s)
Tipificación del Cuerpo , Factor de Crecimiento Nervioso/metabolismo , Rotación , Animales , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Factor de Crecimiento Nervioso/genética , Pruebas de Neutralización , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptor trkA/genética , Receptor trkA/metabolismo , Somitos/efectos de los fármacos , Somitos/embriología
12.
Neurobiol Aging ; 33(4): 833.e1-25, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21958963

RESUMEN

Although amyloid beta (Aß) peptide can promote tau pathology and its toxicity is concurrently tau-dependent, the underlying mechanisms of the in vivo interplay of these proteins remain unsolved. Structural and functional mitochondrial alterations play an early, precipitating role in synaptic failure of Alzheimer's disease (AD) pathogenesis and an aggravated mitochondrial impairment has been described in triple APP/PS/tau transgenic mice carrying both plaques and tangles, if compared with mice overexpressing tau or amyloid precursor protein (APP) alone. Here, we show that a neurotoxic aminoterminal (NH(2))-derived tau fragment mapping between 26 and 230 amino acids of the human tau40 isoform (441 amino acids)-but not the physiological full-length protein-preferentially interacts with Aß peptide(s) in human AD synapses in association with mitochondrial adenine nucleotide translocator-1 (ANT-1) and cyclophilin D. The two peptides-Aß 1-42 and the smaller and more potent NH(2)-26-44 peptide of the longest 20-22 kDa NH(2)-tau fragment-inhibit the ANT-1-dependent adenosine diphosphate-adenosine triphosphate (ADP/ATP) exchange in a noncompetitive and competitive manner, respectively, and together further aggravate the mitochondrial dysfunction by exacerbating the ANT-1 impairment. Taken together, these data establish a common, direct and synergistic toxicity of pathological APP and tau products on synaptic mitochondria and suggest potential, new pathway(s) and target(s) for a combined, more efficient therapeutic intervention of early synaptic dysfunction in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Encéfalo/ultraestructura , Mitocondrias/metabolismo , Neuronas/ultraestructura , Sinaptosomas/patología , Proteínas tau/metabolismo , Translocador 1 del Nucleótido Adenina/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Cerebelo/citología , Ciclofilinas/metabolismo , Homólogo 4 de la Proteína Discs Large , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Neuroblastoma/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Enfermedad de Parkinson/patología , Fragmentos de Péptidos/metabolismo , Ratas , Ratas Wistar , Receptores AMPA/metabolismo , Estaurosporina/farmacología , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/patología , Sinapsis/metabolismo , Sinapsis/patología , Sinaptosomas/metabolismo
13.
PLoS One ; 6(2): e17321, 2011 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-21387003

RESUMEN

During adulthood, the neurotrophin Nerve Growth Factor (NGF) sensitizes nociceptors, thereby increasing the response to noxious stimuli. The relationship between NGF and pain is supported by genetic evidence: mutations in the NGF TrkA receptor in patients affected by an hereditary rare disease (Hereditary Sensory and Autonomic Neuropathy type IV, HSAN IV) determine a congenital form of severe pain insensitivity, with mental retardation, while a mutation in NGFB gene, leading to the aminoacid substitution R100W in mature NGF, determines a similar loss of pain perception, without overt cognitive neurological defects (HSAN V). The R100W mutation provokes a reduced processing of proNGF to mature NGF in cultured cells and a higher percentage of neurotrophin secreted is in the proNGF form. Moreover, using Surface Plasmon Resonance we showed that the R100W mutation does not affect NGF binding to TrkA, while it abolishes NGF binding to p75NTR receptors. However, it remains to be clarified whether the major impact of the mutation is on the biological function of proNGF or of mature NGF and to what extent the effects of the R100W mutation on the HSAN V clinical phenotype are developmental, or whether they reflect an impaired effectiveness of NGF to regulate and mediate nociceptive transmission in adult sensory neurons. Here we show that the R100 mutation selectively alters some of the signaling pathways activated downstream of TrkA NGF receptors. NGFR100 mutants maintain identical neurotrophic and neuroprotective properties in a variety of cell assays, while displaying a significantly reduced pain-inducing activity in vivo (n = 8-10 mice/group). We also show that proNGF has a significantly reduced nociceptive activity, with respect to NGF. Both sets of results jointly contribute to elucidating the mechanisms underlying the clinical HSAN V manifestations, and to clarifying which receptors and intracellular signaling cascades participate in the pain sensitizing action of NGF.


Asunto(s)
Neuropatías Hereditarias Sensoriales y Autónomas/genética , Neuropatías Hereditarias Sensoriales y Autónomas/fisiopatología , Factores de Crecimiento Nervioso/fisiología , Percepción del Dolor/fisiología , Dolor/genética , Sustitución de Aminoácidos/genética , Sustitución de Aminoácidos/fisiología , Animales , Arginina/genética , Células 3T3 BALB , Células Cultivadas , Embrión de Pollo , Predisposición Genética a la Enfermedad , Neuropatías Hereditarias Sensoriales y Autónomas/metabolismo , Humanos , Masculino , Ratones , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Células PC12 , Dolor/metabolismo , Ratas , Receptor de Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/metabolismo , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Triptófano/genética
14.
Proc Natl Acad Sci U S A ; 107(27): 12299-304, 2010 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-20566851

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Factores de Edad , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Factor de Crecimiento Nervioso/deficiencia , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/inmunología , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Unión Proteica , Receptor de Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/inmunología , Receptor trkA/genética , Transducción de Señal , Proteínas tau/metabolismo
15.
Biochem Biophys Res Commun ; 391(1): 824-9, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19945432

RESUMEN

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.


Asunto(s)
Neuropatías Hereditarias Sensoriales y Autónomas/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Insensibilidad Congénita al Dolor/metabolismo , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Neuropatías Hereditarias Sensoriales y Autónomas/genética , Humanos , Mutación , Factor de Crecimiento Nervioso/química , Factor de Crecimiento Nervioso/genética , Insensibilidad Congénita al Dolor/genética , Conformación Proteica , Resonancia por Plasmón de Superficie
16.
Curr Alzheimer Res ; 6(2): 158-70, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19355851

RESUMEN

Nerve growth factor (NGF) deficits are linked to Alzheimer's Disease (AD), due to the role of NGF on basal forebrain cholinergic neurons (BFCN). We have further established that a disequilibrium in NGF signaling and/or processing from its precursor proNGF is also directly and causally related to the aberrant activation of an amyloidogenic route to neurodegeneration. The therapeutic potential of using human NGF to provide a long-lasting cholinergic trophic support, thereby preventing or slowing cognitive decline in AD patients, has therefore a strong rationale. However, a simple and practical means of delivering NGF to the brain in a safe and long-term manner, limiting the undesired adverse effects of NGF in activating nociceptive responses, has represented a significant challenge. For this reason, pilot clinical studies have been performed so far with invasive approaches requiring neurosurgery. We obtained a proof of principle, in neurodegeneration animal models, of an alternative, non-invasive delivery of NGF through an intranasal route, which facilitates access of NGF to the central nervous system (CNS), while minimizing the biodistribution of NGF to compartments where it activates undesired effects, such as pain. The ideal NGF product for a non invasive NGF-based therapy would be a recombinant NGF that, while exhibiting an identical biological activity to that of human NGF, can be traced, against the endogenous NGF, in order to optimize the therapeutical dose range and meet the required therapeutic window. We describe an engineered mutein of hNGF, hNGF-61, that is selectively recognized, against endogenous NGF, by a specific antibody. hNGF-61 mutein has an identical potency and bioactivity profile as hNGF, in vitro and in vivo. Moreover, hNGF-61 and hNGF are equally effective in rescuing the behavioral and neurodegenerative phenotype in adult and aged AD11 anti-NGF mice. Finally, we demonstrated that intranasally delivered hNGF-61 is significantly more effective than ocularly applied hNGF-61, to determine phenotypic rescue in AD11 mice. The development of hNGF-61 towards clinical applications in AD patients is under way.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Factor de Crecimiento Nervioso/uso terapéutico , Enfermedad de Alzheimer/inmunología , Análisis de Varianza , Animales , Anticuerpos/genética , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Modelos Moleculares , Mutación , Factor de Crecimiento Nervioso/farmacología , Pruebas Neuropsicológicas , Ratas , Receptor de Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Alineación de Secuencia , Resonancia por Plasmón de Superficie/métodos , Transfección/métodos
17.
Eur J Neurosci ; 24(5): 1252-64, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16987213

RESUMEN

Alzheimer's disease is a neurodegenerative disorder characterized by neuronal loss associated with a progressive impairment of cognitive functions. Early consequences of Alzheimer's disease include deficit of cholinergic signalling in particular regions controlling memory processes, such as the cortex and hippocampus, and accumulation of beta-amyloid (Abeta) peptide in neuritic plaques. The cholinergic system depends for its integrity and function on nerve growth factor. Chronic nerve growth factor deprivation in transgenic mice (AD11) engineered to produce recombinant neutralizing anti-nerve growth factor antibodies leads to progressive age-dependent Alzheimer's-like neurodegenerative pathology similar to that found in patients with Alzheimer's disease, associated with a selective loss of cholinergic neurones in the basal forebrain. Here we show that in the hippocampus of 6-month-old AD11 mice, Abeta aggregates started appearing in the CA1 region. The accumulation of Abeta was associated with a loss of cholinergic function at CA3-CA1 synapses. Whereas in wild-type mice nicotine induced a persistent increase of synaptic efficacy via alpha7 nicotine acetylcholine receptors, in AD11 mice this alkaloid failed to modify synaptic strength. Moreover, nicotine failed to transiently enhance the frequency of spontaneous miniature glutamatergic currents (miniature excitatory postsynaptic currents) recorded from CA1 but not from CA3 pyramidal neurones of AD11 mice. However, in CA3 principal cells of AD11 mice, the potentiating effect of nicotine on miniature excitatory postsynaptic currents was prevented when Abeta peptide 1-42 was added to the extracellular solution. These data suggest that in AD11 mice, Abeta interferes with nicotine acetylcholine receptors at the level of presynaptic glutamatergic terminals, inhibiting their function possibly through calcium signalling via presynaptic alpha7 nicotine acetylcholine receptors.


Asunto(s)
Hipocampo/citología , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Neuronas/citología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Sinapsis/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Bungarotoxinas/farmacología , Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica/métodos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/farmacología , Inmunohistoquímica/métodos , Técnicas In Vitro , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Antagonistas Nicotínicos/farmacología , Fragmentos de Péptidos/farmacología , Probabilidad , Receptores Nicotínicos/fisiología , Sinapsis/fisiología , Receptor Nicotínico de Acetilcolina alfa 7
18.
Neuroreport ; 13(10): 1369-73, 2002 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-12151805

RESUMEN

In this study we report the expression of TrkA receptor within the rat visual cortex during postnatal development and in adulthood, using a specific monoclonal antibody which recognizes the extracellular domain of TrkA receptor. TrkA was not detected by immunohistochemistry at postnatal day 13 (P13), i.e. before eye opening. At P22 TrkA was mostly localised in cortical fibre-like processes. At P39 and P90, TrkA-positive neuronal cell bodies in supragranular and infragranular layers were found. Using double immunohistochemistry, labelled cells were identified as intrinsic cholinergic neurones, and as interneurones expressing calbindin and neuropeptide Y. We conclude that TrkA is expressed in visual cortical neurones during postnatal development and in adulthood and that its pattern of expression is developmentally regulated.


Asunto(s)
Neuronas/química , Receptor trkA/análisis , Corteza Visual/citología , Factores de Edad , Animales , Anticuerpos Monoclonales , Especificidad de Anticuerpos , Período Crítico Psicológico , Inmunohistoquímica , Ratas , Ratas Wistar , Receptor trkA/inmunología , Corteza Visual/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA