Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Mol Metab ; 6(10): 1321-1329, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29031731

RESUMEN

OBJECTIVE: Pro-opiomelanocortin (POMC)-derived peptides act on neurons expressing the Melanocortin 4 receptor (MC4R) to reduce body weight. Setmelanotide is a highly potent MC4R agonist that leads to weight loss in diet-induced obese animals and in obese individuals with complete POMC deficiency. While POMC deficiency is very rare, 1-5% of severely obese individuals harbor heterozygous mutations in MC4R. We sought to assess the efficacy of Setmelanotide in human MC4R deficiency. METHODS: We studied the effects of Setmelanotide on mutant MC4Rs in cells and the weight loss response to Setmelanotide administration in rodent studies and a human clinical trial. We annotated the functional status of 369 published MC4R variants. RESULTS: In cells, we showed that Setmelanotide is significantly more potent at MC4R than the endogenous ligand alpha-melanocyte stimulating hormone and can disproportionally rescue signaling by a subset of severely impaired MC4R mutants. Wild-type rodents appear more sensitive to Setmelanotide when compared to MC4R heterozygous deficient mice, while MC4R knockout mice fail to respond. In a 28-day Phase 1b clinical trial, Setmelanotide led to weight loss in obese MC4R variant carriers. Patients with POMC defects upstream of MC4R show significantly more weight loss with Setmelanotide than MC4R deficient patients or obese controls. CONCLUSIONS: Setmelanotide led to weight loss in obese people with MC4R deficiency; however, further studies are justified to establish whether Setmelanotide can elicit clinically meaningful weight loss in a subset of the MC4R deficient obese population.


Asunto(s)
Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/deficiencia , alfa-MSH/análogos & derivados , Insuficiencia Suprarrenal/tratamiento farmacológico , Insuficiencia Suprarrenal/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Proopiomelanocortina/deficiencia , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , alfa-MSH/farmacología
2.
Biotechnol Bioeng ; 113(10): 2228-40, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27563853

RESUMEN

Remote activation of specific cells of a heterogeneous population can provide a useful research tool for clinical and therapeutic applications. Here, we demonstrate that photostimulation of gold nanorods (AuNRs) using a tunable near-infrared (NIR) laser at specific longitudinal surface plasmon resonance wavelengths can induce the selective and temporal internalization of calcium in HEK 293T cells. Biotin-PEG-Au nanorods coated with streptavidin Alexa Fluor-633 and biotinylated anti-His antibodies were used to decorate cells genetically modified with His-tagged TRPV1 temperature-sensitive ion channel and AuNRs conjugated to biotinylated RGD peptide were used to decorate integrins in unmodified cells. Plasmonic activation can be stimulated at weak laser power (0.7-4.0 W/cm(2) ) without causing cell damage. Selective activation of TRPV1 channels could be controlled by laser power between 1.0 and 1.5 W/cm(2) . Integrin targeting robustly stimulated calcium signaling due to a dense cellular distribution of nanoparticles. Such an approach represents a functional tool for combinatorial activation of cell signaling in heterogeneous cell populations. Our results suggest that it is possible to induce cell activation via NIR-induced gold nanorod heating through the selective targeting of membrane proteins in unmodified cells to produce calcium signaling and downstream expression of specific genes with significant relevance for both in vitro and therapeutic applications. Biotechnol. Bioeng. 2016;113: 2228-2240. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Oro/efectos de la radiación , Rayos Infrarrojos , Nanotubos/efectos de la radiación , Canales Catiónicos TRPV/metabolismo , Señalización del Calcio/efectos de la radiación , Regulación de la Expresión Génica/fisiología , Regulación de la Expresión Génica/efectos de la radiación , Células HEK293 , Humanos , Nanopartículas del Metal/efectos de la radiación , Dosis de Radiación , Resonancia por Plasmón de Superficie/métodos
3.
Neurobiol Aging ; 44: 159-172, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27318144

RESUMEN

The common apolipoprotein E alleles (ε4, ε3, and ε2) are important genetic risk factors for late-onset Alzheimer's disease, with the ε4 allele increasing risk and reducing the age of onset and the ε2 allele decreasing risk and markedly delaying the age of onset. Preclinical and clinical studies have shown that apolipoprotein E (APOE) genotype also predicts the timing and amount of brain amyloid-ß (Aß) peptide deposition and amyloid burden (ε4 >ε3 >ε2). Using several administration protocols, we now report that direct intracerebral adeno-associated virus (AAV)-mediated delivery of APOE2 markedly reduces brain soluble (including oligomeric) and insoluble Aß levels as well as amyloid burden in 2 mouse models of brain amyloidosis whose pathology is dependent on either the expression of murine Apoe or more importantly on human APOE4. The efficacy of APOE2 to reduce brain Aß burden in either model, however, was highly dependent on brain APOE2 levels and the amount of pre-existing Aß and amyloid deposition. We further demonstrate that a widespread reduction of brain Aß burden can be achieved through a single injection of vector via intrathalamic delivery of AAV expressing APOE2 gene. Our results demonstrate that AAV gene delivery of APOE2 using an AAV vector rescues the detrimental effects of APOE4 on brain amyloid pathology and may represent a viable therapeutic approach for treating or preventing Alzheimer's disease especially if sufficient brain APOE2 levels can be achieved early in the course of the disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E2/genética , Encéfalo/metabolismo , Dependovirus , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Edad de Inicio , Alelos , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Genotipo , Ratones Noqueados , Terapia Molecular Dirigida , Factores de Riesgo
4.
Mol Neurodegener ; 9: 52, 2014 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-25417177

RESUMEN

BACKGROUND: The relationship between the pathogenic amyloid ß-peptide species Aß1-42 and tau pathology has been well studied and suggests that Aß1-42 can accelerate tau pathology in vitro and in vivo. The manners if any in which Aß1-40 interacts with tau remains poorly understood. In order to answer this question, we used cell-based system, transgenic fly and transgenic mice as models to study the interaction between Aß1-42 and Aß1-40. RESULTS: In our established cellular model, live cell imaging (using confocal microscopy) combined with biochemical data showed that exposure to Aß1-42 induced cleavage, phosphorylation and aggregation of wild-type/full length tau while exposure to Aß1-40 didn't. Functional studies with Aß1-40 were carried out in tau-GFP transgenic flies and showed that Aß1-42, as previously reported, disrupted cytoskeletal structure while Aß1-40 had no effect at same dose. To further explore how Aß1-40 affects tau pathology in vivo, P301S mice (tau transgenic mice) were injected intracerebrally with either Aß1-42 or Aß1-40. We found that treatment with Aß1-42 induced tau phosphorylation, cleavage and aggregation of tau in P301S mice. By contrast, Aß1-40 injection didn't alter total tau, phospho-tau (recognized by PHF-1) or cleavage of tau, but interestingly, phosphorylation at Ser262 was shown to be significantly decreased after direct inject of Aß1-40 into the entorhinal cortex of P301S mice. CONCLUSIONS: These results demonstrate that Aß1-40 plays different role in tau pathogenesis compared to Aß1-42. Aß1-40 may have a protective role in tau pathogenesis by reducing phosphorylation at Ser262, which has been shown to be neurotoxic.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Genotipo , Ratones , Microscopía Confocal/métodos , Fragmentos de Péptidos/genética , Fosforilación/fisiología , Proteínas tau/genética
5.
Proc Natl Acad Sci U S A ; 109(38): 15502-7, 2012 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-22927427

RESUMEN

The apolipoprotein E (APOE)-ε4 allele is the strongest genetic risk factor for late-onset, sporadic Alzheimer's disease, likely increasing risk by altering amyloid-ß (Aß) accumulation. We recently demonstrated that the low-density lipoprotein receptor (LDLR) is a major apoE receptor in the brain that strongly regulates amyloid plaque deposition. In the current study, we sought to understand the mechanism by which LDLR regulates Aß accumulation by altering Aß clearance from brain interstitial fluid. We hypothesized that increasing LDLR levels enhances blood-brain barrier-mediated Aß clearance, thus leading to reduced Aß accumulation. Using the brain Aß efflux index method, we found that blood-brain barrier-mediated clearance of exogenously administered Aß is enhanced with LDLR overexpression. We next developed a method to directly assess the elimination of centrally derived, endogenous Aß into the plasma of mice using an anti-Aß antibody that prevents degradation of plasma Aß, allowing its rate of appearance from the brain to be measured. Using this plasma Aß accumulation technique, we found that LDLR overexpression enhances brain-to-blood Aß transport. Together, our results suggest a unique mechanism by which LDLR regulates brain-to-blood Aß clearance, which may serve as a useful therapeutic avenue in targeting Aß clearance from the brain.


Asunto(s)
Amiloidosis/metabolismo , Apolipoproteína E4/genética , Receptores de LDL/biosíntesis , Alelos , Péptidos beta-Amiloides/metabolismo , Animales , Barrera Hematoencefálica , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Insulina/metabolismo , Cinética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Microdiálisis , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA