Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Mol Pharmacol ; 96(2): 138-147, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31189668

RESUMEN

ATP-binding cassette (ABC) transporters such as ABCB1 (P-glycoprotein), ABCC1 (MRP1), and ABCG2 (BCRP) are well known for their role in rendering cancer cells resistant to chemotherapy. Additionally, recent research provided evidence that, along with other ABC transporters (ABCA1 and ABCA7), they might be cornerstones to tackle neurodegenerative diseases. Overcoming chemoresistance in cancer, understanding drug-drug interactions, and developing efficient and specific drugs that alter ABC transporter function are hindered by a lack of in vivo research models, which are fully predictive for humans. Hence, the humanization of ABC transporters in mice has become a major focus in pharmaceutical and neurodegenerative research. Here, we present a characterization of the first Abcc1 humanized mouse line. To preserve endogenous expression profiles, we chose to generate a knockin mouse model that leads to the expression of a chimeric protein that is fully human except for one amino acid. We found robust mRNA and protein expression within all major organs analyzed (brain, lung, spleen, and kidney). Furthermore, we demonstrate the functionality of the expressed human ABCC1 protein in brain and lungs using functional positron emission tomography imaging in vivo. Through the introduction of loxP sites, we additionally enabled this humanized mouse model for highly sophisticated studies involving cell type-specific transporter ablation. Based on our data, the presented mouse model appears to be a promising tool for the investigation of cell-specific ABCC1 function. It can provide a new basis for better translation of preclinical research.


Asunto(s)
Técnicas de Sustitución del Gen/métodos , Pulmón/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Animales , Encéfalo/metabolismo , Humanos , Riñón/metabolismo , Ratones , Ratones Noqueados , Modelos Animales , Tomografía de Emisión de Positrones , Bazo/metabolismo , Distribución Tisular
2.
Brain ; 138(Pt 8): 2370-82, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25991605

RESUMEN

Amyloidosis mouse models of Alzheimer's disease are generally established by transgenic approaches leading to an overexpression of mutated human genes that are known to be involved in the generation of amyloid-ß in Alzheimer's families. Although these models made substantial contributions to the current knowledge about the 'amyloid hypothesis' of Alzheimer's disease, the overproduction of amyloid-ß peptides mimics only inherited (familiar) Alzheimer's disease, which accounts for <1% of all patients with Alzheimer's disease. The inherited form is even regarded a 'rare' disease according to the regulations for funding of the European Union (www.erare.eu). Here, we show that mice that are double-deficient for neprilysin (encoded by Mme), one major amyloid-ß-degrading enzyme, and the ABC transporter ABCC1, a major contributor to amyloid-ß clearance from the brain, develop various aspects of sporadic Alzheimer's disease mimicking the clinical stage of mild cognitive impairment. Using behavioural tests, electrophysiology and morphological analyses, we compared different ABC transporter-deficient animals and found that alterations are most prominent in neprilysin × ABCC1 double-deficient mice. We show that these mice have a reduced probability to survive, show increased anxiety in new environments, and have a reduced working memory performance. Furthermore, we detected morphological changes in the hippocampus and amygdala, e.g. astrogliosis and reduced numbers of synapses, leading to defective long-term potentiation in functional measurements. Compared to human, murine amyloid-ß is poorly aggregating, due to changes in three amino acids at N-terminal positions 5, 10, and 13. Interestingly, our findings account for the action of early occurring amyloid-ß species/aggregates, i.e. monomers and small amyloid-ß oligomers. Thus, neprilysin × ABCC1 double-deficient mice present a new model for early effects of amyloid-ß-related mild cognitive impairment that allows investigations without artificial overexpression of inherited Alzheimer's disease genes.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Neprilisina/genética , Enfermedad de Alzheimer/metabolismo , Animales , Disfunción Cognitiva/genética , Modelos Animales de Enfermedad , Potenciación a Largo Plazo , Ratones Noqueados , Neprilisina/metabolismo , Neuronas/metabolismo
3.
J Alzheimers Dis ; 77(3): 1209-1221, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32831204

RESUMEN

BACKGROUND: The recent failure of clinical trials to treat Alzheimer's disease (AD) indicates that the current approach of modifying disease is either wrong or is too late to be efficient. Mild cognitive impairment (MCI) denotes the phase between the preclinical phase and clinical overt dementia. AD mouse models that overexpress human amyloid-ß (Aß) are used to study disease pathogenesis and to conduct drug development/testing. However, there is no direct correlation between the Aß deposition, the age of onset, and the severity of cognitive dysfunction. OBJECTIVE: To detect and predict MCI when Aß plaques start to appear in the hippocampus of an AD mouse. METHODS: We trained wild-type and AD mice in a Morris water maze (WM) task with different inter-trial intervals (ITI) at 3 months of age and assessed their WM performance. Additionally, we used a classification algorithm to predict the genotype (APPtg versus wild-type) of an individual mouse from their respective WM data. RESULTS: MCI can be empirically detected using a short-ITI protocol. We show that the ITI modulates the spatial learning of AD mice without affecting the formation of spatial memory. Finally, a simple classification algorithm such as logistic regression on WM data can give an accurate prediction of the cognitive dysfunction of a specific mouse. CONCLUSION: MCI can be detected as well as predicted simultaneously with the onset of Aß deposition in the hippocampus in AD mouse model. The mild cognitive impairment prediction can be used for assessing the efficacy of a treatment.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/psicología , Disfunción Cognitiva/genética , Disfunción Cognitiva/psicología , Enfermedad de Alzheimer/patología , Animales , Disfunción Cognitiva/patología , Femenino , Predicción , Humanos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
4.
Acta Neuropathol Commun ; 5(1): 49, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28637503

RESUMEN

Amyloid-ß (Aß) deposition is one of the hallmarks of the amyloid hypothesis in Alzheimer's disease (AD). Mouse models using APP-transgene overexpression to generate amyloid plaques have shown to model only certain parts of the disease. The extent to which the data from mice can be transferred to man remains controversial. Several studies have shown convincing treatment results in reducing Aß and enhancing cognition in mice but failed totally in human. One model-dependent factor has so far been almost completely neglected: the endogenous expression of mouse APP and its effects on the transgenic models and the readout for therapeutic approaches.Here, we report that hAPP-transgenic models of amyloidosis devoid of endogenous mouse APP expression (mAPP-knockout / mAPPko) show increased amounts and higher speed of Aß deposition than controls with mAPP. The number of senile plaques and the level of aggregated hAß were elevated in mAPPko mice, while the deposition in cortical blood vessels was delayed, indicating an alteration in the general aggregation propensity of hAß together with endogenous mAß. Furthermore, the cellular response to Aß deposition was modulated: mAPPko mice developed a pronounced and age-dependent astrogliosis, while microglial association to amyloid plaques was diminished. The expression of human and murine aggregation-prone proteins with differing amino acid sequences within the same mouse model might not only alter the extent of deposition but also modulate the route of pathogenesis, and thus, decisively influence the study outcome, especially in translational research.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/metabolismo , Fragmentos de Péptidos/metabolismo , Precursor de Proteína beta-Amiloide/genética , Amiloidosis/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Caspasas/metabolismo , Femenino , Humanos , Masculino , Meninges/irrigación sanguínea , Meninges/metabolismo , Meninges/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Neuronas/patología , Placa Amiloide/metabolismo , Placa Amiloide/patología
5.
Acta Neuropathol Commun ; 4(1): 91, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27566602

RESUMEN

Alzheimer's disease primarily occurs as sporadic disease and is accompanied with vast socio-economic problems. The mandatory basic research relies on robust and reliable disease models to overcome increasing incidence and emerging social challenges. Rodent models are most efficient, versatile, and predominantly used in research. However, only highly artificial and mostly genetically modified models are available. As these 'engineered' models reproduce only isolated features, researchers demand more suitable models of sporadic neurodegenerative diseases. One very promising animal model was the South American rodent Octodon degus, which was repeatedly described as natural 'sporadic Alzheimer's disease model' with 'Alzheimer's disease-like neuropathology'. To unveil advantages over the 'artificial' mouse models, we re-evaluated the age-dependent, neurohistological changes in young and aged Octodon degus (1 to 5-years-old) bred in a wild-type colony in Germany. In our hands, extensive neuropathological analyses of young and aged animals revealed normal age-related cortical changes without obvious signs for extensive degeneration as seen in patients with dementia. Neither significant neuronal loss nor enhanced microglial activation were observed in aged animals. Silver impregnation methods, conventional, and immunohistological stains as well as biochemical fractionations revealed neither amyloid accumulation nor tangle formation. Phosphoepitope-specific antibodies against tau species displayed similar intraneuronal reactivity in both, young and aged Octodon degus.In contrast to previous results, our study suggests that Octodon degus born and bred in captivity do not inevitably develop cortical amyloidosis, tangle formation or neuronal loss as seen in Alzheimer's disease patients or transgenic disease models.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Octodon , Envejecimiento/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones Transgénicos , Octodon/metabolismo , Proteínas tau/metabolismo
6.
Mol Neurobiol ; 53(7): 4728-44, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26319560

RESUMEN

Parkinson's disease and dementia with Lewy bodies are major challenges in research and clinical medicine world-wide and contribute to the most common neurodegenerative disorders. Previously, specific mitochondrial polymorphisms have been found to enhance clearance of amyloid-ß from the brain of APP-transgenic mice leading to beneficial clinical outcome. It has been discussed whether specific mitochondrial alterations contribute to disease progression or even prevent toxic peptide deposition, as seen in many neurodegenerative diseases. Here, we investigated α-synuclein-transgenic C57BL/6J mice with the A30P mutation, and a novel A30P C57BL/6J mouse model with three mitochondrial DNA polymorphisms in the ND3, COX3 and mtRNA(Arg) genes, as found in the inbred NOD/LtJ mouse strain. We were able to detect that the new model has increased mitochondrial complex II-respiration which occurs in parallel to neuronal loss and improved motor performance, although it exhibits higher amounts of high molecular weight species of α-synuclein. High molecular weight aggregates of different peptides are controversially discussed in the light of neurodegeneration. A favourable hypothesis states that high molecular weight species are protective and of minor importance for the pathogenesis of neurodegenerative disorders as compared to the extreme neurotoxic monomers and oligomers. Summarising, our results point to a potentially protective and beneficial effect of specific mitochondrial polymorphisms which cause improved mitochondrial complex II-respiration in α-synucleinopathies, an effect that could be exploited further for pharmaceutical interventions.


Asunto(s)
Complejo II de Transporte de Electrones/metabolismo , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Respiración de la Célula/fisiología , Complejo II de Transporte de Electrones/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Mitocondrias/genética , Mitocondrias/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , alfa-Sinucleína/genética
7.
Acta Neuropathol Commun ; 4: 25, 2016 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-26984535

RESUMEN

INTRODUCTION: Alzheimer's disease (AD) is associated with the accumulation of ß-amyloid (Aß) as senile plaques in the brain, thus leading to neurodegeneration and cognitive impairment. Plaque formation depends not merely on the amount of generated Aß peptides, but more importantly on their effective removal. Chronic infections with neurotropic pathogens, most prominently the parasite Toxoplasma (T.) gondii, are frequent in the elderly, and it has been suggested that the resulting neuroinflammation may influence the course of AD. In the present study, we investigated how chronic T. gondii infection and resulting neuroinflammation affect plaque deposition and removal in a mouse model of AD. RESULTS: Chronic infection with T. gondii was associated with reduced Aß and plaque load in 5xFAD mice. Upon infection, myeloid-derived CCR2(hi) Ly6C(hi) monocytes, CCR2(+) Ly6C(int), and CCR2(+) Ly6C(low) mononuclear cells were recruited to the brain of mice. Compared to microglia, these recruited mononuclear cells showed highly increased phagocytic capacity of Aß ex vivo. The F4/80(+) Ly6C(low) macrophages expressed high levels of Triggering Receptor Expressed on Myeloid cells 2 (TREM2), CD36, and Scavenger Receptor A1 (SCARA1), indicating phagocytic activity. Importantly, selective ablation of CCR2(+) Ly6C(hi) monocytes resulted in an increased amount of Aß in infected mice. Elevated insulin-degrading enzyme (IDE), matrix metalloproteinase 9 (MMP9), as well as immunoproteasome subunits ß1i/LMP2, ß2i/MECL-1, and ß5i/LMP7 mRNA levels in the infected brains indicated increased proteolytic Aß degradation. Particularly, LMP7 was highly expressed by the recruited mononuclear cells in the brain, suggesting a novel mechanism of Aß clearance. CONCLUSIONS: Our results indicate that chronic Toxoplasma infection ameliorates ß-amyloidosis in a murine model of AD by activation of the immune system, specifically by recruitment of Ly6C(hi) monocytes and by enhancement of phagocytosis and degradation of soluble Aß. Our findings provide evidence for a modulatory role of inflammation-induced Aß phagocytosis and degradation by newly recruited peripheral immune cells in the pathophysiology of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Macrófagos/metabolismo , Monocitos/fisiología , Fagocitosis/fisiología , Toxoplasmosis/metabolismo , Toxoplasmosis/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Anticuerpos/farmacología , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Encéfalo/parasitología , Encéfalo/patología , Proteínas de Unión al Calcio/metabolismo , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/patología , Fagocitosis/genética , Placa Amiloide/metabolismo , Placa Amiloide/patología , Receptores CCR2/genética , Receptores CCR2/inmunología
8.
Arch Med Res ; 45(8): 738-43, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25446622

RESUMEN

In elderly subjects, depression and dementia often coincide but the actual reason is currently unknown. Does a causal link exist or is it just a reactive effect of the knowledge to suffer from dementia? The ABC transporter superfamily may represent a causal link between these mental disorders. Since the transporters ABCB1 and ABCC1 have been discovered as major ß-amyloid-exporting molecules at the blood-brain barrier and ABCC1 was found to be directly activated by St. John's wort (SJW), depression and dementia certainly share an important pathophysiologic link. It was recognized that herbal anti-depressant formulations made from SJW are at least as effective for the treatment of unipolar depression in old age as classical pharmacotherapy, while having fewer side effects (Cochrane reports, 2008). SJW is known to activate various metabolizing and transport systems in the body, with cytochrome P450 enzymes and ABC transporters being most important. Does the treatment of depression in elderly subjects using pharmacological compounds or phytomedical extracts target a mechanism that also accounts for peptide storage in Alzheimer's disease and perhaps other proteopathies of the brain? In this review we summarize recent data that point to a common mechanism and present the first promising causal treatment results of demented elderly subjects with distinct SJW extracts. Insufficient trans-barrier clearance may indeed present a common problem in all the proteopathies of the brain where toxic peptides are deposited in a location-specific manner. Thus, activation of efflux molecules holds promise for future treatment of this large group of devastating disorders.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Barrera Hematoencefálica/metabolismo , Depresión/fisiopatología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Anciano , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Transporte Biológico , Depresión/tratamiento farmacológico , Humanos , Hypericum/química , Masculino , Preparaciones de Plantas/uso terapéutico
9.
Mech Ageing Dev ; 134(10): 506-15, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24012632

RESUMEN

Ageing is the main risk factor for the development of dementing neurodegenerative diseases (NDs) and it is accompanied by the accumulation of variations in mitochondrial DNA. The resulting tissue-specific alterations in ATP production and availability cause deteriorations of cerebral clearance mechanisms that are important for the removal of toxic peptides and its aggregates. ABC transporters were shown to be the most important exporter superfamily for toxic peptides, e.g. ß-amyloid and α-synuclein. Their activity is highly dependent on the availability of ATP and forms a directed energy-exporter network, linking decreased mitochondrial function with highly impaired ABC transporter activity and disease progression. In this paper, we describe a network based on interactions between ageing, energy metabolism, regeneration, accumulation of toxic peptides and the development of proteopathies of the brain with a focus on Alzheimer's disease (AD). Additionally, we provide new experimental evidence for interactions within this network in regenerative processes in AD.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Envejecimiento/genética , Envejecimiento/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/patología , Metabolismo Energético/genética , Humanos , Ratones , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
10.
Eur J Microbiol Immunol (Bp) ; 3(1): 21-27, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23814667

RESUMEN

Alzheimer's disease (AD) is by far the most common neurodegenerative disease. AD is histologically characterized not only by extracellular senile plaques and vascular deposits consisting of ß-amyloid (Aß) but also by accompanying neuroinflammatory processes involving the brain's microglia. The importance of the microglia is still in controversial discussion, which currently favors a protective function in disease progression. Recent findings by different research groups highlighted the importance of strain-specific and mitochondria-specific genomic variations in mouse models of cerebral ß-amyloidosis. Here, we want to summarize our previously presented data and add new results that draw attention towards the consideration of strain-specific genomic alterations in the setting of APP transgenes. We present data from APP-transgenic mice in commonly used C57Bl/6J and FVB/N genomic backgrounds and show a direct influence on the kinetics of Aß deposition and the activity of resident microglia. Plaque size, plaque deposition rate and the total amount of Aß are highest in C57Bl/6J mice as compared to the FVB/N genomic background, which can be explained at least partially by a reduced microglia activity towards amyloid deposits in the C57BL/6J strain.

11.
PLoS One ; 7(4): e35613, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22545122

RESUMEN

BACKGROUND: ATP-binding cassette (ABC) transporters are essential regulators of organismic homeostasis, and are particularly important in protecting the body from potentially harmful exogenous substances. Recently, an increasing number of in vitro observations have indicated a functional role of ABC transporters in the differentiation and maintenance of stem cells. Therefore, we sought to determine brain-related phenotypic changes in animals lacking the expression of distinct ABC transporters (ABCB1, ABCG2 or ABCC1). METHODOLOGY AND PRINCIPAL FINDINGS: Analyzing adult neurogenesis in ABC transporter-deficient animals in vivo and neuronal stem/progenitor cells in vitro resulted in complex findings. In vivo, the differentiation of neuronal progenitors was hindered in ABC transporter-deficient mice (ABCB1(0/0)) as evidenced by lowered numbers of doublecortin(+) (-36%) and calretinin(+) (-37%) cells. In vitro, we confirmed that this finding is not connected to the functional loss of single neural stem/progenitor cells (NSPCs). Furthermore, assessment of activity, exploratory behavior, and anxiety levels revealed behavioral alterations in ABCB1(0/0) and ABCC1(0/0) mice, whereas ABCG2(0/0) mice were mostly unaffected. CONCLUSION AND SIGNIFICANCE: Our data show that single ABC transporter-deficiency does not necessarily impair neuronal progenitor homeostasis on the single NSPC level, as suggested by previous studies. However, loss of distinct ABC transporters impacts global brain homeostasis with far ranging consequences, leading to impaired neurogenic functions in vivo and even to distinct behavioral phenotypes. In addition to the known role of ABC transporters in proteopathies such as Parkinson's disease and Alzheimer's disease, our data highlight the importance of understanding the general function of ABC transporters for the brain's homeostasis and the regeneration potential.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Neurogénesis , Neuronas/citología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Ansiedad/genética , Ansiedad/metabolismo , Conducta Animal , Encéfalo/citología , Encéfalo/metabolismo , Proliferación Celular , Células Cultivadas , Femenino , Eliminación de Gen , Masculino , Ratones , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Neuronas/metabolismo , Células Madre/citología , Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA