Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Mol Neurosci ; 17: 1379743, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38756707

RESUMEN

Hearing loss constitutes a major global health concern impacting approximately 1.5 billion people worldwide. Its incidence is undergoing a substantial surge with some projecting that by 2050, a quarter of the global population will experience varying degrees of hearing deficiency. Environmental factors such as aging, exposure to loud noise, and the intake of ototoxic medications are implicated in the onset of acquired hearing loss. Ototoxicity resulting in inner ear damage is a leading cause of acquired hearing loss worldwide. This could be minimized or avoided by early testing of hearing functions in the preclinical phase of drug development. While the assessment of ototoxicity is well defined for drug candidates in the hearing field - required for drugs that are administered by the otic route and expected to reach the middle or inner ear during clinical use - ototoxicity testing is not required for all other therapeutic areas. Unfortunately, this has resulted in more than 200 ototoxic marketed medications. The aim of this publication is to raise awareness of drug-induced ototoxicity and to formulate some recommendations based on available guidelines and own experience. Ototoxicity testing programs should be adapted to the type of therapy, its indication (targeting the ear or part of other medications classes being potentially ototoxic), and the number of assets to test. For multiple molecules and/or multiple doses, screening options are available: in vitro (otic cell assays), ex vivo (cochlear explant), and in vivo (in zebrafish). In assessing the ototoxicity of a candidate drug, it is good practice to compare its ototoxicity to that of a well-known control drug of a similar class. Screening assays provide a streamlined and rapid method to know whether a drug is generally safe for inner ear structures. Mammalian animal models provide a more detailed characterization of drug ototoxicity, with a possibility to localize and quantify the damage using functional, behavioral, and morphological read-outs. Complementary histological measures are routinely conducted notably to quantify hair cells loss with cochleogram. Ototoxicity studies can be performed in rodents (mice, rats), guinea pigs and large species. However, in undertaking, or at the very least attempting, all preclinical investigations within the same species, is crucial. This encompasses starting with pharmacokinetics and pharmacology efficacy studies and extending through to toxicity studies. In life read-outs include Auditory Brainstem Response (ABR) and Distortion Product OtoAcoustic Emissions (DPOAE) measurements that assess the activity and integrity of sensory cells and the auditory nerve, reflecting sensorineural hearing loss. Accurate, reproducible, and high throughput ABR measures are fundamental to the quality and success of these preclinical trials. As in humans, in vivo otoscopic evaluations are routinely carried out to observe the tympanic membrane and auditory canal. This is often done to detect signs of inflammation. The cochlea is a tonotopic structure. Hair cell responsiveness is position and frequency dependent, with hair cells located close to the cochlea apex transducing low frequencies and those at the base transducing high frequencies. The cochleogram aims to quantify hair cells all along the cochlea and consequently determine hair cell loss related to specific frequencies. This measure is then correlated with the ABR & DPOAE results. Ototoxicity assessments evaluate the impact of drug candidates on the auditory and vestibular systems, de-risk hearing loss and balance disorders, define a safe dose, and optimize therapeutic benefits. These types of studies can be initiated during early development of a therapeutic solution, with ABR and otoscopic evaluations. Depending on the mechanism of action of the compound, studies can include DPOAE and cochleogram. Later in the development, a GLP (Good Laboratory Practice) ototoxicity study may be required based on otic related route of administration, target, or known potential otic toxicity.

2.
J Acoust Soc Am ; 146(5): 3743, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31795705

RESUMEN

Guinea pigs have been used in diverse studies to better understand acquired hearing loss induced by noise and ototoxic drugs. The guinea pig has its best hearing at slightly higher frequencies relative to humans, but its hearing is more similar to humans than the rat or mouse. Like other rodents, it is more vulnerable to noise injury than the human or nonhuman primate models. There is a wealth of information on auditory function and vulnerability of the inner ear to diverse insults in the guinea pig. With respect to the assessment of potential otoprotective agents, guinea pigs are also docile animals that are relatively easy to dose via systemic injections or gavage. Of interest, the cochlea and the round window are easily accessible, notably for direct cochlear therapy, as in the chinchilla, making the guinea pig a most relevant and suitable model for hearing. This article reviews the use of the guinea pig in basic auditory research, provides detailed discussion of its use in studies on noise injury and other injuries leading to acquired sensorineural hearing loss, and lists some therapeutics assessed in these laboratory animal models to prevent acquired sensorineural hearing loss.


Asunto(s)
Modelos Animales de Enfermedad , Cobayas/fisiología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Estimulación Acústica/métodos , Experimentación Animal/normas , Animales , Pérdida Auditiva Provocada por Ruido/etiología , Pérdida Auditiva Provocada por Ruido/prevención & control , Especificidad de la Especie
3.
Brain Behav Immun ; 80: 697-710, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31100368

RESUMEN

In adult brain, the chemokine CXCL12 and its receptors CXCR4 and CXCR7 are expressed in neural progenitor and glial cells. Conditional Cxcl12 or Cxcr4 gene knockout in mice leads to severe alterations in neural progenitor proliferation, migration and differentiation. As adult hippocampal neurogenesis is involved in learning and memory processes, we investigated the long-term effects of reduced expression of CXCL12 or CXCR7 in heterozygous Cxcl12+/- and Cxcr7+/- animals (KD mice) on hippocampal neurogenesis, neuronal differentiation and memory processing. In Cxcl12 KD mice, Cxcr4 mRNA expression was reduced, whereas Cxcr7 was slightly increased. Conversely, in Cxcr7 KD mice, both Cxcr4 and Cxcl12 mRNA levels were decreased. Moreover, Cxcl12 KD animals showed marked behavioral and learning deficits that were associated with impaired neurogenesis in the hippocampus. Conversely, Cxcr7 KD animals showed mild learning deficits with normal neurogenesis, but reduced cell differentiation, measured with doublecortin immunolabeling. These findings suggested that a single Cxcl12 or Cxcr7 allele might not be sufficient to maintain the hippocampal niche functionality throughout life, and that heterozygosity might represent a susceptibility factor for memory dysfunction progression.


Asunto(s)
Quimiocina CXCL12/metabolismo , Aprendizaje/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Receptores CXCR/metabolismo , Animales , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Quimiocina CXCL12/genética , Quimiocinas/metabolismo , Femenino , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurogénesis/fisiología , Neuroglía/fisiología , Receptores CXCR/genética , Transducción de Señal
4.
Aging Dis ; 9(4): 664-673, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30090654

RESUMEN

Age-related hearing loss (ARHL) is the most common sensory disorder in the elderly population. SAMP8 mouse model presents accelerated senescence and has been identified as a model of gerontological research. SAMP8 displays a progressive age-related decline in brain function associated with a progressive hearing loss mimicking human aging memory deficits and ARHL. The molecular mechanisms associated with SAMP8 senescence process involve oxidative stress leading to chronic inflammation and apoptosis. Here, we studied the effect of N-acetylcysteine (NAC), an antioxidant, on SAMP8 hearing loss and memory to determine the potential interest of this model in the study of new antioxidant therapies. We observed a strong decrease of auditory brainstem response thresholds from 45 to 75 days of age and an increase of distortion product amplitudes from 60 to 75 days in NAC treated group compared to vehicle. Moreover, NAC treated group presented also an increase of memory performance at 60 and 105 days of age. These results confirm that NAC delays the senescence process by slowing the age-related hearing loss, protecting the cochlear hair cells and improving memory, suggesting that antioxidants could be a pharmacological target for age-related hearing and memory loss.

5.
J Psychopharmacol ; 32(2): 174-190, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29215943

RESUMEN

RATIONALE: The prevalence of Alzheimer's disease is increased in people with Down syndrome. The pathology appears much earlier than in the general population, suggesting a predisposition to develop Alzheimer's disease. Down syndrome results from trisomy of human chromosome 21, leading to overexpression of possible Alzheimer's disease candidate genes, such as amyloid precursor protein gene. To better understand how the Down syndrome context results in increased vulnerability to Alzheimer's disease, we analysed amyloid-ß [25-35] peptide toxicity in the Tc1 mouse model of Down syndrome, in which ~75% of protein coding genes are functionally trisomic but, importantly, not amyloid precursor protein. RESULTS: Intracerebroventricular injection of oligomeric amyloid-ß [25-35] peptide in three-month-old wildtype mice induced learning deficits, oxidative stress, synaptic marker alterations, activation of glycogen synthase kinase-3ß, inhibition of protein kinase B (AKT), and apoptotic pathways as compared to scrambled peptide-treated wildtype mice. Scrambled peptide-treated Tc1 mice presented high levels of toxicity markers as compared to wildtype mice. Amyloid-ß [25-35] peptide injection in Tc1 mice induced significant learning deficits and enhanced glycogen synthase kinase-3ß activity in the cortex and expression of apoptotic markers in the hippocampus and cortex. Interestingly, several markers, including oxidative stress, synaptic markers, glycogen synthase kinase-3ß activity in the hippocampus and AKT activity in the hippocampus and cortex, were unaffected by amyloid-ß [25-35] peptide injection in Tc1 mice. CONCLUSIONS: Tc1 mice present several toxicity markers similar to those observed in amyloid-ß [25-35] peptide-treated wildtype mice, suggesting that developmental modifications in these mice modify their response to amyloid peptide. However, amyloid toxicity led to severe memory deficits in this Down syndrome mouse model.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/toxicidad , Síndrome de Down/fisiopatología , Trastornos de la Memoria/fisiopatología , Fragmentos de Péptidos/toxicidad , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/administración & dosificación , Animales , Biomarcadores/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Síndrome de Down/complicaciones , Síndrome de Down/genética , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipocampo/metabolismo , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Fragmentos de Péptidos/administración & dosificación , Índice de Severidad de la Enfermedad
6.
Eur Neuropsychopharmacol ; 25(11): 2170-82, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26381812

RESUMEN

Dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs) and cdc2-like kinases (CLKs) are implicated in the onset and progression of Down syndrome (DS) and Alzheimer's disease (AD). DYRK1A has emerged as a possible link between amyloid-ß (Aß) and Tau, the major pathological proteins in AD. We here assessed the neuroprotective potential of a novel inhibitor of DYRKs/CLKs. The Leucettine L41, acting preferentially on DYRK1A, was tested in Aß25-35-treated mice, a nontransgenic model of AD-like toxicity. We co-injected intracerebroventricularly oligomeric Aß25-35 peptide and L41 in Swiss male mice. After 7 days, they were submitted to behavioral tests addressing spatial and non-spatial, short- and long-term memories. The oxidative stress, apoptotic markers, kinases involved in Tau phosphorylation, and synaptic integrity were analyzed by Western blot and ELISA in the hippocampus. L41, tested at 0.4, 1.2, 4 µg, prevented the Aß25-35-induced memory deficits in the Y-maze, passive avoidance and water-maze tests, with the most active dose being 4 µg. The inhibitor prevented the Aß25-35-induced oxidative stress, as revealed by measures of lipid peroxidation levels and reactive oxygen species accumulation, and abolished Aß25-35-induced expression of pro-apoptotic markers. L41 prevented the Aß25-35-induced decrease of AKT activation and increase of glycogen synthase kinase-3ß (GSK-3ß) activation, resulting in a decrease of Tau phosphorylation. Finally, L41 restored Aß25-35-reduced levels of synaptic markers. The novel DYRK1A-preferential inhibitor L41 therefore prevented Aß25-35-induced memory impairments and neurotoxicity in the mouse hippocampus. These in vivo data highlighted particularly DYRK1A as a major kinase involved in Aß pathology and suggested therapeutic developments for DYRK1A inhibitors in AD.


Asunto(s)
Dioxoles/farmacología , Imidazoles/farmacología , Trastornos de la Memoria/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Péptidos beta-Amiloides , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Hipocampo/patología , Masculino , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/patología , Ratones , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Fragmentos de Péptidos , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/enzimología , Sinapsis/patología , Proteínas tau/metabolismo , Quinasas DyrK
7.
J Psychopharmacol ; 27(11): 1044-57, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23813967

RESUMEN

Erythropoietin (EPO) promotes neurogenesis and neuroprotection. We here compared the protection induced by two EPO formulations in a rodent model of Alzheimer's disease (AD): rHu-EPO and a low sialic form, Neuro-EPO. We used the intracerebroventricular administration of aggregated Aß25₋35 peptide, a non-transgenic AD model. rHu-EPO was tested at 125-500 µg/kg intraperitoneally and Neuro-EPO at 62-250 µg/kg intranasally (IN). Behavioural procedures included spontaneous alternation, passive avoidance, water-maze and object recognition, to address spatial and non-spatial, short- and long-term memories. Biochemical markers of Aß25₋35 toxicity in the mouse hippocampus were examined and cell loss in the CA1 layer was determined. rHu-EPO and Neuro-EPO led to a significant prevention of Aß25₋35-induced learning deficits. Both EPO formulations prevented the induction of lipid peroxidation in the hippocampus, showing an antioxidant activity. rHu-EPO (250 µg/kg) or Neuro-EPO (125 µg/kg) prevented the Aß25₋35-induced increase in Bax level, TNFα and IL-1ß production and decrease in Akt activation. A significant prevention of the Aß25₋35-induced cell loss in CA1 was also observed. EPO is neuroprotective in the Aß25₋35 AD model, confirming its potential as an endogenous neuroprotection system that could be boosted for therapeutic efficacy. We here identified a new IN formulation of EPO showing high neuroprotective activity. Considering its efficacy, ease and safety, IN Neuro-EPO is a new promising therapeutic agent in AD.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/antagonistas & inhibidores , Química Farmacéutica , Modelos Animales de Enfermedad , Eritropoyetina/administración & dosificación , Eritropoyetina/farmacología , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/antagonistas & inhibidores , Administración Intranasal , Péptidos beta-Amiloides/toxicidad , Animales , Recuento de Células , Relación Dosis-Respuesta a Droga , Eritropoyetina/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Interleucina-1beta/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Receptores de Eritropoyetina/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Retención en Psicología/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
8.
J Mol Cell Biol ; 5(5): 284-93, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23892208

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by intracellular neurofibrillary tangle formation and extracellular amyloid-ß (Aß) deposition. To date, microglia seem to act as double-edged swords, being either beneficial (e.g. clearance of Aß) or detrimental (e.g. secretion of neurotoxic factors) in AD. Following a rather intense debate on the question, a consensus has emerged that microglia can renew themselves via proliferation of already differentiated microglia as well as via the de novo recruitment of monocytes of mouse models of AD. However, recent advances suggest distinct function for resident and bone marrow-derived microglia (BMDM), and have emphasized the neuroprotective functions of BMDM. BMDM is the only subset of cells that restrict cerebral amyloidosis in the AD brain, which has been recently attributed to CCR2(+) monocytes. Moreover, an impaired recruitment of CCR2(+) monocytes has been reported in AD patients, as seen from the CCR2(+) monocytopenia found in the bloodstream and BM. The present review summarizes the current knowledge on the roles and dysfunctions of CCR2(+) monocytes in AD and their potential as key therapeutic targets.


Asunto(s)
Enfermedad de Alzheimer/patología , Monocitos/patología , Receptores CCR2/deficiencia , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Quimiocina CCL2/metabolismo , Humanos , Microglía/metabolismo , Microglía/patología , Monocitos/metabolismo , Receptores CCR2/metabolismo
9.
Neuropsychopharmacology ; 38(9): 1706-23, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23493042

RESUMEN

The main objective of the present study was to establish whether the mixed σ1/muscarinic ligand ANAVEX2-73, shown to be neuroprotective in Alzheimer's disease (AD) models in vivo and currently in clinical phase I/IIa, could have the ability to reduce the appearance of hyperphosphorylated Tau and amyloid-ß1₋42 (Aß1₋42 in the Aß25₋35 mouse model of AD. We therefore first confirmed that Aß25₋35 injection induced hyperphosphorylation of Tau protein, by showing that it rapidly decreased Akt activity and activated glycogen synthase kinase-3ß (GSK-3ß) in the mouse hippocampus. Second, we showed that the kinase activation, and resulting Tau alteration, directly contributed to the amyloid toxicity, as co-administration of the selective GSK-3ß inhibitor 2-thio(3-iodobenzyl)-5-(1-pyridyl)-[1,3,4]-oxidiazole blocked both Tau phosphorylation and Aß25₋35-induced memory impairments. Third, we analyzed the ANAVEX2-73 effect on Tau phosphorylation and activation of the related kinase pathways (Akt and GSK-3ß). And fourth, we also addressed the impact of the drug on Aß25₋35-induced Aß1₋42 seeding and observed that the compound significantly blocked the increase in Aß1₋42 and C99 levels in the hippocampus, suggesting that it may alleviate amyloid load in AD models. The comparison with PRE-084, a selective and reference σ1 receptor agonist, and xanomeline, a muscarinic ligand presenting similar profile as ANAVEX2-73 on M1 and M2 subtypes, confirmed that both muscarinic and σ1 targets are involved in the ANAVEX2-73 effects. The drug, acting synergistically on both targets, but with moderate affinity, presents a promising pharmacological profile.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/biosíntesis , Furanos/farmacología , Agonistas Muscarínicos/farmacología , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/biosíntesis , Receptores sigma/agonistas , Proteínas tau/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/administración & dosificación , Péptidos beta-Amiloides/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Furanos/uso terapéutico , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Infusiones Intraventriculares , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Ratones , Morfolinas/farmacología , Morfolinas/uso terapéutico , Agonistas Muscarínicos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Oxadiazoles/administración & dosificación , Oxadiazoles/farmacología , Oxadiazoles/uso terapéutico , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/administración & dosificación , Piridinas/farmacología , Piridinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Tiadiazoles/farmacología , Tiadiazoles/uso terapéutico
10.
Front Cell Neurosci ; 6: 51, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23125823

RESUMEN

Alzheimer's disease (AD) is characterized by a progressive memory decline and numerous pathological abnormalities, including amyloid ß (Aß) accumulation in the brain and synaptic dysfunction. Here we wanted to study whether these brain changes were associated with alteration in the population of monocyte subsets since accumulating evidence supports the concept that the innate immune system plays a role in the etiology of this disease. We then determined the immune profile together with expression of genes encoding synaptic proteins and neurotrophins in APP(Swe)/PS1 mice and their age-matched wild-type (WT) littermates. We found that the progressive cognitive decline and the dramatic decrease in the expression of numerous synaptic markers and neurotrophins correlated with a major defect in the subset of circulating inflammatory monocytes. Indeed the number of CX(3)CR1(low)Ly6-C(high)CCR2(+)Gr1(+) monocytes remained essentially similar between 5 weeks and 6 months of age in APP(Swe)/PS1 mice, while these cells significantly increased in 6-month-old WT littermates. Of great interest is that the onset of cognitive decline was closely associated with the accumulation of soluble Aß, disruption of synaptic activity, alteration in the BDNF system, and a defective production in the subset of CX(3)CR1(low)Ly6-C(high)CCR2(+)Gr1(+) monocytes. However, these memory impairments can be prevented or restored by boosting the monocytic production, using a short treatment of macrophage colony-stimulating factor (M-CSF). In conclusion, low CCR2(+) monocyte production by the hematopoietic system may be a direct biomarker of the cognitive decline in a context of AD.

11.
Mol Med ; 18: 297-313, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22160221

RESUMEN

Monocytes emigrate from bone marrow, can infiltrate into brain, differentiate into microglia and clear amyloid ß (Aß) from the brain of mouse models of Alzheimer's disease (AD). Here we show that these mechanisms specifically require CC-chemokine receptor 2 (CCR2) expression in bone marrow cells (BMCs). Disease progression was exacerbated in APP(Swe)/PS1 mice (transgenic mice expressing a chimeric amyloid precursor protein [APPSwe] and human presenilin 1 [PS1]) harboring CCR2-deficient BMCs. Indeed, transplantation of CCR2-deficient BMCs enhanced the mnesic deficit and increased the amount of soluble Aß and expression of transforming growth factor (TGF)-ß1 and TGF-ß receptors. By contrast, transplantation of wild-type bone marrow stem cells restored memory capacities and diminished soluble Aß accumulation in APP(Swe)/PS1 and APP(Swe)/PS1/CCR2⁻/⁻ mice. Finally, gene therapy using a lentivirus-expressing CCR2 transgene in BMCs prevented cognitive decline in this mouse model of AD. Injection of CCR2 lentiviruses restored CCR2 expression and functions in monocytes. The presence of these cells in the brain of non-irradiated APP(Swe)/PS1/CCR2⁻/⁻ mice supports the concept that they can be used as gene vehicles for AD. Decreased CCR2 expression in bone marrow-derived microglia may therefore play a major role in the etiology of this neurodegenerative disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Trastornos del Conocimiento/metabolismo , Células Madre Hematopoyéticas/metabolismo , Receptores CCR2/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Reacción de Prevención , Encéfalo/metabolismo , Encéfalo/patología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Expresión Génica , Masculino , Memoria , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta1/genética
12.
Am J Pathol ; 179(1): 315-34, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21703413

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative pathology characterized by the presence of senile plaques and neurofibrillary tangles, accompanied by synaptic and neuronal loss. The major component of senile plaques is an amyloid ß protein (Aß) formed by pathological processing of the Aß precursor protein. We assessed the time-course and regional effects of a single intracerebroventricular injection of aggregated Aß fragment 25-35 (Aß(25-35)) in rats. Using a combined biochemical, behavioral, and morphological approach, we analyzed the peptide effects after 1, 2, and 3 weeks in the hippocampus, cortex, amygdala, and hypothalamus. The scrambled Aß(25-35) peptide was used as negative control. The aggregated forms of Aß peptides were first characterized using electron microscopy, infrared spectroscopy, and Congo Red staining. Intracerebroventricular injection of Aß(25-35) decreased body weight, induced short- and long-term memory impairments, increased endocrine stress, cerebral oxidative and cellular stress, neuroinflammation, and neuroprotective reactions, and modified endogenous amyloid processing, with specific time-course and regional responses. Moreover, Aß(25-35), the presence of which was shown in the different brain structures and over 3 weeks, provoked a rapid glial activation, acetylcholine homeostasis perturbation, and hippocampal morphological alterations. In conclusion, the acute intracerebroventricular Aß(25-35) injection induced substantial central modifications in rats, highly reminiscent of the human physiopathology, that could contribute to physiological and cognitive deficits observed in AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Encéfalo/efectos de los fármacos , Inflamación/etiología , Ovillos Neurofibrilares/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Acetilcolina/metabolismo , Animales , Encéfalo/citología , Corteza Cerebral/efectos de los fármacos , Humanos , Inflamación/patología , Masculino , Memoria a Largo Plazo/efectos de los fármacos , Ovillos Neurofibrilares/patología , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Espectroscopía Infrarroja por Transformada de Fourier
13.
J Neurosci ; 31(16): 6208-20, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21508244

RESUMEN

Circulating monocytoid cells have the ability to infiltrate nervous tissue, differentiate into microglia, and clear amyloid-ß (Aß) from the brain of mouse models of Alzheimer's disease. Interaction between the chemokine CCL2 and its CC chemokine receptor 2 (CCR2) plays a critical role in the recruitment of inflammatory monocytes into the injured/diseased brain. Here, we show that CCR2 deficiency aggravates mnesic deficits and amyloid pathology in transgenic mice expressing the chimeric mouse/human ß-amyloid precursor protein and presenilin 1 (APP(Swe)/PS1). Indeed, memory impairment was accelerated and enhanced in APP(Swe)/PS1/CCR2(-/-) mice. Apparition of cognitive decline occurred earlier (i.e., at 3 months of age before plaque formation) and correlated with intracellular accumulation of soluble oligomeric forms of Aß. Memory deficits worsened with age and were aggravated in APP(Swe)/PS1/CCR2(-/-) mice compared with their respective control groups. Soluble Aß assemblies increased significantly in APP(Swe)/PS1 mice in a context of CCR2 deficiency, whereas the plaque load remained relatively similar in the brain of aging APP(Swe)/PS1 and APP(Swe)/PS1/CCR2(-/-) mice. However, CCR2 deficiency stimulated the expression of TGF-ß1, TGF-ß receptors, and CX(3)CR1 transcripts in plaque-associated microglia, a pattern that is characteristic of an antiinflammatory subset of myeloid cells. A decreased expression of CCR2 could play a potential role in the etiology of Alzheimer's disease, a neurodegenerative pathology that could be treated by a genetic upregulation of the transgene in monocytoid cells.


Asunto(s)
Enfermedad de Alzheimer/genética , Encéfalo/patología , Trastornos del Conocimiento/genética , Placa Amiloide/genética , Receptores CCR2/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Encéfalo/metabolismo , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Inmunohistoquímica , Hibridación in Situ , Modelos Lineales , Masculino , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Ratones , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Placa Amiloide/patología , Presenilina-1/genética , Presenilina-1/metabolismo , Receptores CCR2/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
14.
Mol Cell Neurosci ; 46(1): 55-66, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20708081

RESUMEN

Depression is potentially life-threatening. The most important neuroendocrine abnormality in this disorder is hypothalamo-pituitary-adrenocortical (HPA) axis hyperactivity. Recent findings suggest that all depression treatments may boost the neurotrophin production especially brain-derived neurotrophic factor (BDNF). Moreover, BDNF is highly involved in the regulation of HPA axis activity. The aim of this study was to determine the impact of chronic stress (restraint 3h/day for 3 weeks) on animal behavior and HPA axis activity in parallel with hippocampus, hypothalamus and pituitary BDNF levels. Chronic stress induced changes in anxiety (light/dark box test) and anhedonic states (sucrose preference test) and in depressive-like behavior (forced swimming test); general locomotor activity and body temperature were modified and animal body weight gain was reduced by 17%. HPA axis activity was highly modified by chronic stress, since basal levels of mRNA and peptide hypothalamic contents in CRH and AVP and plasma concentrations in ACTH and corticosterone were significantly increased. The HPA axis response to novel acute stress was also modified in chronically stressed rats, suggesting adaptive mechanisms. Basal BDNF contents were increased in the hippocampus, hypothalamus and pituitary in chronically stressed rats and the BDNF response to novel acute stress was also modified. This multiparametric study showed that chronic restraint stress induced a depressive-like state that was sustained by mechanisms associated with BDNF regulation.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Depresión/fisiopatología , Sistema Hipotálamo-Hipofisario/fisiología , Sistema Hipófiso-Suprarrenal/fisiología , Restricción Física/psicología , Estrés Psicológico/fisiopatología , Animales , Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/genética , Masculino , Actividad Motora/fisiología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/psicología
15.
J Innate Immun ; 1(5): 480-93, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20375605

RESUMEN

Toll-like receptors (TLRs) are essential to mount a rapid innate immune reaction to pathogens. Although TLR2 is the key receptor for pathogen-associated molecular patterns from Gram-positive bacteria, a robust transcriptional activation of the gene encoding this receptor takes place in the brain of mice exposed to the TLR4 ligand lipopolysaccharide (LPS). TLR2 gene expression is actually used as a reliable marker of activated microglia in vivo, but its functions remain unknown. The present study investigated the role of this receptor in mediating LPS-induced gene expression in the mouse brain. Immune genes were measured using both in situ hybridization and real time RT-PCR. Despite the robust microglial TLR2 expression, this receptor does not modulate transcriptional activity by TLR4 signaling. TLR2-deficient mice and their wild-type littermates had similar IkappaBalpha mRNA levels and induction of innate immune genes from 6 h to 10 days after LPS injection. In contrast, NF-kappaB activity, cytokine, chemokine, TLR2 and CD14 transcripts were no longer detected in MyD88-deficient mice. Indeed, the hybridization signal for most of the transcripts measured in this study was similar in the brain of MyD88(-/-) mice exposed to either saline or LPS. These data indicate that while TLR2 transcription is dependent on MyD88 signaling in microglia, this innate immune receptor is not involved in the immune response to LPS. On the other hand, MyD88 pathway is essential for the endotoxin to induce expression of immune genes in the central nervous system.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica , Microglía/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inmunidad Innata , Inflamación/inmunología , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , FN-kappa B/metabolismo , Receptor Toll-Like 2/genética
16.
Neuropsychopharmacology ; 33(7): 1584-602, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17805310

RESUMEN

Chromatin remodeling by posttranslational modification of histones plays an important role in brain plasticity, including memory, response to stress and depression. The importance of H3/4 histones acetylation by CREB-binding protein (CBP) or related histone acetyltransferase, including p300, was specifically demonstrated using knockout (KO) mouse models. The physiological role of a related protein that also acts as a transcriptional coactivator with intrinsic histone acetylase activity, the p300/CBP-associated factor (PCAF), is poorly documented. We analyzed the behavioral phenotype of homozygous male and female PCAF KO mice and report a marked impact of PCAF deletion on memory processes and stress response. PCAF KO animals showed short-term memory deficits at 2 months of age, measured using spontaneous alternation, object recognition, or acquisition of a daily changing platform position in the water maze. Acquisition of a fixed platform location was delayed, but preserved, and no passive avoidance deficit was noted. No gender-related difference was observed. These deficits were associated with hippocampal alterations in pyramidal cell layer organization, basal levels of Fos immunoreactivity, and MAP kinase activation. PCAF KO mice also showed an exaggerated response to acute stress, forced swimming, and conditioned fear, associated with increased plasma corticosterone levels. Moreover, learning and memory impairments worsened at 6 and 12 months of age, when animals failed to acquire the fixed platform location in the water maze and showed passive avoidance deficits. These observations demonstrate that PCAF histone acetylase is involved lifelong in the chromatin remodeling necessary for memory formation and response to stress.


Asunto(s)
Memoria/fisiología , Estrés Psicológico/genética , Estrés Psicológico/fisiopatología , Factores de Transcripción p300-CBP/deficiencia , Factores de Edad , Análisis de Varianza , Animales , Reacción de Prevención/fisiología , Conducta Animal , Condicionamiento Psicológico/fisiología , Corticosterona/sangre , Modelos Animales de Enfermedad , Emociones/fisiología , Conducta Exploratoria/fisiología , Miedo , Femenino , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/genética , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Noqueados , Reconocimiento Visual de Modelos/fisiología , Factores Sexuales , Estrés Psicológico/patología
17.
Psychoneuroendocrinology ; 32(8-10): 1062-78, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17928160

RESUMEN

Depression is characterized by hypothalamo-pituitary-adrenocortical (HPA) axis hyperactivity. In this major mood disorder, neurosteroids and neurotrophins, particularly brain-derived neurotrophic factor (BDNF), seem to be implicated and have some antidepressant effects. BDNF is highly involved in regulation of the HPA axis, whereas neurosteroids effects have never been clearly established. In this systematic in vivo study, we showed that the principal neuroactive steroids, namely dehydroepiandrosterone (DHEA), pregnenolone (PREG) and their sulfate esters (DHEA-S and PREG-S), along with allopregnanolone (ALLO), stimulated HPA axis activity, while also modulating central BDNF contents. In detail, DHEA, DHEA-S, PREG, PREG-S and ALLO induced corticotropin-releasing hormone (CRH) and/or arginine vasopressin (AVP) synthesis and release at the hypothalamic level, thus enhancing plasma adrenocorticotropin hormone (ACTH) and corticosterone (CORT) concentrations. This stimulation of the HPA axis occurred concomitantly with BDNF modifications at the hippocampus, amygdala and hypothalamus levels. We showed that these neurosteroids induced rapid effects, probably via neurotransmitter receptors and delayed effects perhaps after metabolization in other neuroactive steroids. We highlighted that they had peripheral effects directly at the adrenal level by inducing CORT release, certainly after estrogenic metabolization. In addition, we showed that, at the dose used, only DHEA, DHEA-S and PREG-S had antidepressant effects. In conclusion, these results highly suggest that part of the HPA axis and antidepressant effects of neuroactive steroids could be mediated by BDNF, particularly at the amygdala level. They also suggest that neurosteroids effects on central BDNF could partially explain the trophic properties of these molecules.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Neuropéptidos/farmacología , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Esteroides/farmacología , Animales , Encéfalo/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Deshidroepiandrosterona/administración & dosificación , Sulfato de Deshidroepiandrosterona/administración & dosificación , Inyecciones Intraperitoneales , Masculino , Neuropéptidos/administración & dosificación , Pregnanolona/administración & dosificación , Pregnenolona/administración & dosificación , Ratas , Ratas Sprague-Dawley , Esteroides/administración & dosificación
18.
J Endocrinol ; 188(3): 425-33, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16522723

RESUMEN

Brain-derived neurotrophic factor (BDNF) has been extensively studied in the central nervous system as a survival and differentiation factor and in plasticity processes. In vitro, BDNF has been shown to stimulate cellular differentiation and neurohormones synthesis and release. We demonstrated that BDNF is a potent and specific stimulatory agent of somatostatin (SRIH) synthesis in primary cultures of hypothalamic neurons. However, less information is available about its function on SRIH neurons in vivo. In the present study, we examined the effect of in vivo intracerebroventricular BDNF administration in adult non-anesthetized male rats. Two distinct experimental approaches were used: acute intracerebroventricular injection and long-term (14 days) continuous infusion (Alzet micro-pumps). We demonstrate that single intracerebroventricular BDNF injections (5 microg/rat) induce an early (60 and 180 min) decrease in the SRIH mRNA signal in the hypothalamic periventricular nucleus (PeVN) accompanied by a decrease of the hypothalamic SRIH content. 48 h after the acute injection, SRIH mRNA levels and peptide content strongly and significantly increased. After continuous intracerebroventricular BDNF administration (12 microg/day for 14 days), a significant increase in the SRIH hypothalamic content was observed. Nevertheless, the increase in peptide content was not correlated with a similar increase in the PeVN messenger level. These findings show the involvement of BDNF in the in vivo regulation of somatostatinergic neurons in adult rats, which clearly differs according to the BDNF administration mode.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Hipotálamo/química , ARN Mensajero/análisis , Somatostatina/genética , Animales , Relación Dosis-Respuesta a Droga , Hipotálamo/efectos de los fármacos , Procesamiento de Imagen Asistido por Computador , Hibridación in Situ/métodos , Bombas de Infusión , Inyecciones Intraventriculares , Masculino , Núcleo Hipotalámico Paraventricular/química , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Somatostatina/análisis , Estimulación Química , Factores de Tiempo
19.
Mol Cell Neurosci ; 27(3): 280-95, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15519243

RESUMEN

Immobilization stress induces in adult male rats rapid activation of brain derived neurotrophic factor (BDNF) expression in the hypothalamic paraventricular nucleus (PVN) preceding the increases in corticotropin releasing hormone (CRH) and arginin-vasopressin (AVP) expression. The BDNF mRNA signal belatedly co-localizes with CRH and AVP mRNA signals in the PVN, as determined by in situ hybridization. Intracerebroventricular BDNF injections (5 microg/rat) in non-anesthetized adult male rats induce a gradual increase in the CRH mRNA signal whereas AVP mRNA signal progressively decreases in the parvocellular and magnocellular PVN portions. At the same time, the CRH hypothalamic content decreases while the AVP content increases. These variations are accompanied by increases in ACTH and corticosterone plasma concentrations. These results strongly suggest that BDNF could be a stress-responsive intercellular messenger since when it is exogenously administered acts as an important and early component in the activation and recruitment of hypothalamic CRH and AVP neurons.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Arginina Vasopresina/biosíntesis , Corticosterona/sangre , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Masculino , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Estrés Fisiológico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...