Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Cell Neurosci ; 17: 1179796, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37346371

RESUMEN

While motor and cortical neurons are affected in C9orf72 amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD), it remains largely unknown if and how non-neuronal cells induce or exacerbate neuronal damage. We differentiated C9orf72 ALS/FTD patient-derived induced pluripotent stem cells into microglia (iPSC-MG) and examined their intrinsic phenotypes. Similar to iPSC motor neurons, C9orf72 ALS/FTD iPSC-MG mono-cultures form G4C2 repeat RNA foci, exhibit reduced C9orf72 protein levels, and generate dipeptide repeat proteins. Healthy control and C9orf72 ALS/FTD iPSC-MG equally express microglial specific genes and perform microglial functions, including inflammatory cytokine release and phagocytosis of extracellular cargos, such as synthetic amyloid beta peptides and healthy human brain synaptoneurosomes. RNA sequencing analysis revealed select transcriptional changes of genes associated with neuroinflammation or neurodegeneration in diseased microglia yet no significant differentially expressed microglial-enriched genes. Moderate molecular and functional differences were observed in C9orf72 iPSC-MG mono-cultures despite the presence of C9orf72 pathological features suggesting that a diseased microenvironment may be required to induce phenotypic changes in microglial cells and the associated neuronal dysfunction seen in C9orf72 ALS/FTD neurodegeneration.

3.
Acta Neuropathol ; 138(1): 49-65, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30945056

RESUMEN

The hexanucleotide repeat expansion GGGGCC (G4C2)n in the C9orf72 gene is the most common genetic abnormality associated with amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Recent findings suggest that dysfunction of nuclear-cytoplasmic trafficking could affect the transport of RNA binding proteins in C9orf72 ALS/FTD. Here, we provide evidence that the RNA editing enzyme adenosine deaminase acting on RNA 2 (ADAR2) is mislocalized in C9orf72 repeat expansion mediated ALS/FTD. ADAR2 is responsible for adenosine (A) to inosine (I) editing of double-stranded RNA, and its function has been shown to be essential for survival. Here we show the mislocalization of ADAR2 in human induced pluripotent stem cell-derived motor neurons (hiPSC-MNs) from C9orf72 patients, in mice expressing (G4C2)149, and in C9orf72 ALS/FTD patient postmortem tissue. As a consequence of this mislocalization we observe alterations in RNA editing in our model systems and across multiple brain regions. Analysis of editing at 408,580 known RNA editing sites indicates that there are vast RNA A to I editing aberrations in C9orf72-mediated ALS/FTD. These RNA editing aberrations are found in many cellular pathways, such as the ALS pathway and the crucial EIF2 signaling pathway. Our findings suggest that the mislocalization of ADAR2 in C9orf72 mediated ALS/FTD is responsible for the alteration of RNA processing events that may impact vast cellular functions, including the integrated stress response (ISR) and protein translation.


Asunto(s)
Adenosina Desaminasa/genética , Proteína C9orf72/genética , Edición de ARN/genética , Proteínas de Unión al ARN/genética , Esclerosis Amiotrófica Lateral/genética , Animales , Expansión de las Repeticiones de ADN/genética , Demencia Frontotemporal/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones Transgénicos , Enfermedad de Pick/genética
4.
J Liposome Res ; 28(2): 97-105, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28103719

RESUMEN

Light chain (AL) amyloidosis is a disease associated with significant morbidity and mortality arising from multi-organ injury induced by amyloidogenic light chain proteins (LC). There is no available treatment to reverse the toxicity of LC. We previously showed that chaperone glycoprotein clusterin (CLU) and nanoliposomes (NL), separately, restore human microvascular endothelial function impaired by LC. In this work, we aim to prepare PEGylated-nanoliposomal clusterin (NL-CLU) formulations that could allow combined benefit against LC while potentially enabling efficient delivery to microvascular tissue, and test efficacy on human arteriole endothelial function. NL-CLU was prepared by a conjugation reaction between the carboxylated surface of NL and the primary amines of the CLU protein. NL were made of phosphatidylcholine (PC), cholesterol (Chol) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-2000] (DSPE-PEG 2000 carboxylic acid) at 70:25:5 mol%. The protective effect of NL-CLU was tested by measuring the dilation response to acetylcholine and papaverine in human adipose arterioles exposed to LC. LC treatment significantly reduced the dilation response to acetylcholine and papaverine; co-treatment of LC with PEGylated-nanoliposomal CLU or free CLU restored the dilator response. NL-CLU is a feasible and promising approach to reverse LC-induced endothelial damage.


Asunto(s)
Proteínas Amiloidogénicas/metabolismo , Amiloidosis/tratamiento farmacológico , Clusterina/administración & dosificación , Endotelio Vascular/efectos de los fármacos , Liposomas/química , Nanopartículas/química , Acetilcolina/química , Arteriolas/efectos de los fármacos , Arteriolas/metabolismo , Colesterol/química , Clusterina/química , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Papaverina/química , Tamaño de la Partícula , Fosfatidilcolinas/química , Polietilenglicoles/química , Vasodilatación/efectos de los fármacos
5.
J Am Heart Assoc ; 5(6)2016 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-27412900

RESUMEN

BACKGROUND: Light chain amyloidosis (AL) is associated with high mortality, especially in patients with advanced cardiovascular involvement. It is caused by toxicity of misfolded light chain proteins (LC) in vascular, cardiac, and other tissues. There is no treatment to reverse LC tissue toxicity. We tested the hypothesis that nanoliposomes composed of monosialoganglioside, phosphatidylcholine, and cholesterol (GM1 ganglioside-containing nanoliposomes [NLGM1]) can protect against LC-induced human microvascular dysfunction and assess mechanisms behind the protective effect. METHODS AND RESULTS: The dilator responses of ex vivo abdominal adipose arterioles from human participants without AL to acetylcholine and papaverine were measured before and after exposure to LC (20 µg/mL) with or without NLGM1 (1:10 ratio for LC:NLGM1 mass). Human umbilical vein endothelial cells were exposed for 18 to 20 hours to vehicle, LC with or without NLGM1, or NLGM1 and compared for oxidative and nitrative stress response and cellular viability. LC impaired arteriole dilator response to acetylcholine, which was restored by co-treatment with NLGM1. LC decreased endothelial cell nitric oxide production and cell viability while increasing superoxide and peroxynitrite; these adverse effects were reversed by NLGM1. NLGM1 increased endothelial cell protein expression of antioxidant enzymes heme oxygenase 1 and NAD(P)H quinone dehydrogenase 1 and increased nuclear factor, erythroid 2 like 2 (Nrf-2) protein. Nrf-2 gene knockdown reduced antioxidant stress response and reversed the protective effects of NLGM1. CONCLUSIONS: NLGM1 protects against LC-induced human microvascular endothelial dysfunction through increased nitric oxide bioavailability and reduced oxidative and nitrative stress mediated by Nrf-2-dependent antioxidant stress response. These findings point to a potential novel therapeutic approach for light chain amyloidosis.


Asunto(s)
Colesterol/administración & dosificación , Endotelio Vascular/efectos de los fármacos , Gangliósidos/administración & dosificación , Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas/complicaciones , Fosfatidilcolinas/administración & dosificación , Enfermedades Vasculares/prevención & control , Tejido Adiposo/irrigación sanguínea , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Supervivencia Celular/fisiología , Combinación de Medicamentos , Células Endoteliales/metabolismo , Técnicas de Silenciamiento del Gen/métodos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas/prevención & control , Masculino , Persona de Mediana Edad , NAD(P)H Deshidrogenasa (Quinona)/genética , Factor 2 Relacionado con NF-E2/genética , Nanopartículas/administración & dosificación , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Papaverina/farmacología , Ácido Peroxinitroso/biosíntesis , Interferencia de ARN/fisiología , ARN Interferente Pequeño/fisiología , Especies Reactivas de Oxígeno/metabolismo , Superóxidos/metabolismo , Transfección , Enfermedades Vasculares/fisiopatología , Vasodilatadores/farmacología
6.
J Cereb Blood Flow Metab ; 36(2): 405-12, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26661197

RESUMEN

We tested whether nanoliposomes containing phosphatidylcholine, cholesterol and phosphatidic acid (NLPA) prevent ß-amyloid 1-42 (Aß42) fibrillation and Aß42-induced human arteriole endothelial dysfunction. NLPA abolished Aß42 fibril formation (thioflavin-T fluorescence/electron microscopy). In ex-vivo human adipose and leptomeningeal arterioles, Aß42 impaired dilator response to acetylcholine that was reversed by NLPA; this protection was abolished by L-NG-nitroarginine methyl ester. Aß42 reduced human umbilical vein endothelial cell NO production that was restored by NLPA. Nanoliposomes prevented Aß42 amyloid formation, reversed Aß42-induced human microvascular endothelial dysfunction and may be useful in Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides , Arteriolas/patología , Endotelio Vascular/patología , Liposomas/uso terapéutico , Fragmentos de Péptidos , Enfermedades Vasculares/inducido químicamente , Enfermedades Vasculares/prevención & control , Acetilcolina/antagonistas & inhibidores , Acetilcolina/farmacología , Tejido Adiposo/irrigación sanguínea , Colesterol/administración & dosificación , Colesterol/uso terapéutico , Humanos , Técnicas In Vitro , Masculino , Meninges/irrigación sanguínea , Persona de Mediana Edad , NG-Nitroarginina Metil Éster/farmacología , Nanopartículas/uso terapéutico , Óxido Nítrico/biosíntesis , Ácidos Fosfatidicos/administración & dosificación , Ácidos Fosfatidicos/uso terapéutico , Fosfatidilcolinas/administración & dosificación , Fosfatidilcolinas/uso terapéutico , Enfermedades Vasculares/patología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
7.
Diab Vasc Dis Res ; 12(3): 154-63, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25852133

RESUMEN

Cardiovascular (CV) disease is the leading cause of mortality and morbidity in patients with type 2 diabetes mellitus (T2DM). However, improving glycaemic control alone has not decreased CV events. Therapies that improve glycaemic control, CV disease risk factors and CV function are more likely to be successful. Dipeptidyl peptidase-4 (DPP-4) inhibitors prevent breakdown of incretin hormones glucagon-like peptide-1(GLP-1) and glucose-dependent insulinotropic peptide and improve glycaemic control in patients with T2DM. DPP-4 acts on other substrates, many associated with cardioprotection. Thus, inhibition of DPP-4 may lead to elevations in these potentially beneficial substrates. Data from animal studies and small observational studies in humans suggest that DPP-4 inhibitors may potentially reduce CV risk. However, recently completed CV outcome trials in patients with T2DM and CV disease or at high risk of adverse CV events have shown that the DPP-4 inhibitors saxagliptin and alogliptin neither increased nor decreased major adverse CV events.


Asunto(s)
Glucemia/efectos de los fármacos , Enfermedades Cardiovasculares/prevención & control , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Incretinas/uso terapéutico , Adamantano/análogos & derivados , Adamantano/uso terapéutico , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Enfermedades Cardiovasculares/etiología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/enzimología , Dipéptidos/uso terapéutico , Inhibidores de la Dipeptidil-Peptidasa IV/efectos adversos , Humanos , Incretinas/efectos adversos , Piperidinas/uso terapéutico , Factores Protectores , Medición de Riesgo , Factores de Riesgo , Resultado del Tratamiento , Uracilo/análogos & derivados , Uracilo/uso terapéutico
8.
Diabetes ; 64(7): 2624-35, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25720388

RESUMEN

GLP-1 receptor (GLP-1R) agonists may improve endothelial function (EF) via metabolic improvement and direct vascular action. The current study determined the effect of GLP-1R agonist exenatide on postprandial EF in type 2 diabetes and the mechanisms underlying GLP-1R agonist-mediated vasodilation. Two crossover studies were conducted: 36 participants with type 2 diabetes received subcutaneous exenatide or placebo for 11 days and EF, and glucose and lipid responses to breakfast and lunch were determined; and 32 participants with impaired glucose tolerance (IGT) or diet-controlled type 2 diabetes had EF measured before and after intravenous exenatide, with or without the GLP-1R antagonist exendin-9. Mechanisms of GLP-1R agonist action were studied ex vivo on human subcutaneous adipose tissue arterioles and endothelial cells. Subcutaneous exenatide increased postprandial EF independent of reductions in plasma glucose and triglycerides. Intravenous exenatide increased fasting EF, and exendin-9 abolished this effect. Exenatide elicited eNOS activation and NO production in endothelial cells, and induced dose-dependent vasorelaxation and reduced high-glucose or lipid-induced endothelial dysfunction in arterioles ex vivo. These effects were reduced with AMPK inhibition. In conclusion, exenatide augmented postprandial EF in subjects with diabetes and prevented high-glucose and lipid-induced endothelial dysfunction in human arterioles. These effects were largely direct, via GLP-1R and AMPK activation.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Péptidos/farmacología , Receptores de Glucagón/agonistas , Vasodilatación/efectos de los fármacos , Ponzoñas/farmacología , Proteínas Quinasas Activadas por AMP/fisiología , Glucemia/análisis , Células Cultivadas , Estudios Cruzados , Diabetes Mellitus Tipo 2/fisiopatología , Método Doble Ciego , Células Endoteliales/fisiología , Exenatida , Femenino , Receptor del Péptido 1 Similar al Glucagón , Humanos , Masculino , Receptores de Glucagón/fisiología , Triglicéridos/sangre
9.
J Neurosci Methods ; 235: 123-9, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25004204

RESUMEN

BACKGROUND: Evidence point to vascular dysfunction and hypoperfusion as early abnormalities in Alzheimer's disease (AD); probing their mechanistic bases can lead to new therapeutic approaches. We tested the hypotheses that ß-amyloid peptide induces endothelial dysfunction and oxidative stress in human microvasculature and that response will be similar between peripheral adipose and brain leptomeningeal arterioles. NEW METHOD: Abdominal subcutaneous arterioles from living human subjects (n=17) and cadaver leptomeningeal arterioles (n=6) from rapid autopsy were exposed to Aß1-42 (Aß) for 1-h and dilation response to acetylcholine/papaverine were measured and compared to baseline response. Adipose arteriole reactive oxygen species (ROS) production and nitrotyrosine content were measured. COMPARISON WITH EXISTING METHODS: Methods described allow direct investigation of human microvessel functional response that cannot be replicated by human noninvasive imaging or post-mortem histology. RESULTS: Adipose arterioles exposed to 2 µM Aß showed impaired dilation to acetylcholine that was reversed by antioxidant polyethylene glycol superoxide dismutase (PEG-SOD) (Aß-60.9 ± 6%, control-93.2 ± 1.8%, Aß+PEGSOD-84.7 ± 3.9%, both p<0.05 vs. Aß). Aß caused reduced dilation to papaverine. Aß increased adipose arteriole ROS production and increased arteriole nitrotyrosine content. Leptomeningeal arterioles showed similar impaired response to acetylcholine when exposed to Aß (43.0 ± 6.2% versus 81.1 ± 5.7% control, p<0.05). CONCLUSION: Aß exposure induced adipose arteriole endothelial and non-endothelial dysfunction and oxidative stress that were reversed by antioxidant treatment. Aß-induced endothelial dysfunction was similar between peripheral adipose and leptomeningeal arterioles. Ex vivo living adipose and cadaver leptomeningeal arterioles are viable, novel and practical human tissue models to study Alzheimer's vascular pathophysiology.


Asunto(s)
Tejido Adiposo/irrigación sanguínea , Péptidos beta-Amiloides , Arteriolas/fisiopatología , Células Endoteliales/fisiología , Meninges/irrigación sanguínea , Fragmentos de Péptidos , Abdomen/irrigación sanguínea , Acetilcolina/farmacología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estrés Oxidativo/fisiología , Papaverina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Vasodilatadores/farmacología
10.
J Liposome Res ; 24(1): 69-73, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24236475

RESUMEN

CONTEXT: A newly-recognized pathogenic mechanism underlying light chain amyloidosis (AL) involves endothelial dysfunction and cell injury caused by misfolded light chain proteins (LC). Nanoliposomes (NL) are artificial phospholipid vesicles that could attach to misfolded proteins and reduce tissue injury. OBJECTIVE: To test whether co-treatment with NL reduces LC-induced endothelial dysfunction and cell death. METHODS: Abdominal subcutaneous adipose arterioles from 14 non-AL subjects were cannulated; dilator response to acetylcholine and papaverine were measured at baseline and following 1-hour exposure to LC (20 µg/mL, 2 purified from AL subjects' urine, 1 from human recombinant LC [AL-09]) ± NL (phosphatidylcholine/cholesterol/phosphatidic acid 70/25/5 molar ratio) or NL alone. Human aortic artery endothelial cells (HAEC) were exposed to Oregon Green-labeled LC ± NL for 24 hours and intracellular LC and apoptosis (Hoechst stain) were measured. Circular dichroism spectroscopy was performed on AL-09 LC ± NL to follow changes in secondary structure and protein thermal stability. RESULTS: LC caused impaired dilation to acetylcholine that was restored by NL (control - 94.0 ± 1.8%, LC - 65.0 ± 7.1%, LC + NL - 95.3 ± 1.8%, p ≤ 0.001 LC versus control or LC + NL). NL protection was inhibited by L-NG-nitroarginine methyl ester. NL increased the beta sheet structure of LC, reduced endothelial cell internalization of LC and protected against LC-induced endothelial cell death. CONCLUSIONS: LC induced human adipose arteriole endothelial dysfunction and endothelial cell death, which were reversed by co-treatment with NL. This protection may partly be due to enhancing LC protein structure and reducing LC internalization. Nanoliposomes represent a promising new class of agents to ameliorate tissue injury from protein misfolding diseases such as AL.


Asunto(s)
Amiloide/química , Amiloidosis/tratamiento farmacológico , Endotelio/efectos de los fármacos , Liposomas/uso terapéutico , Nanopartículas/uso terapéutico , Anciano , Apoptosis/efectos de los fármacos , Endotelio/lesiones , Endotelio/patología , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/patología , Humanos , Liposomas/química , Masculino , Persona de Mediana Edad , Nanopartículas/química , Deficiencias en la Proteostasis/tratamiento farmacológico
11.
Atherosclerosis ; 225(1): 220-3, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22981431

RESUMEN

UNLABELLED: Misfolded immunoglobulin light chain proteins (LC) in light chain amyloidosis (AL) are toxic to vascular tissues. We tested the hypothesis that chaperone protein clusterin preserves endothelial function and cell survival during LC exposure. METHODS: LC (20 µg/mL) were given to human aortic endothelial cells (EC) for 24-h and clusterin protein/gene expression and secretion were measured. DNA fragmentation was measured with/without recombinant clusterin (Clu, 300 ng/mL). Adipose arterioles (non-AL subjects) were tested for dilator responses to acetylcholine/papaverine at baseline and after 1-h of LC ± Clu. RESULTS: LC reduced EC clusterin secretion, protein and gene expression while increasing DNA fragmentation. Clu attenuated LC-induced DNA fragmentation and restored dilator response to acetylcholine (logEC50: control -7.05 ± 0.2, LC + Clu -6.53 ± 0.4, LC -4.28 ± 0.7, p < 0.05 versus control, LC + Clu). CONCLUSIONS: LC induced endothelial cell death and dysfunction while reducing clusterin protein/gene expression and secretion. Exogenous clusterin attenuated LC toxicity. This represents a new pathobiologic mechanism and therapeutic target for AL amyloidosis.


Asunto(s)
Amiloidosis/fisiopatología , Clusterina/fisiología , Células Endoteliales/fisiología , Cadenas Ligeras de Inmunoglobulina , Arteriolas/fisiopatología , Muerte Celular/efectos de los fármacos , Clusterina/biosíntesis , Fragmentación del ADN/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Cadenas Ligeras de Inmunoglobulina/metabolismo , Cadenas Ligeras de Inmunoglobulina/farmacología , Masculino , Persona de Mediana Edad , Vasodilatación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...