Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 18802, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37914766

ABSTRACT

Medin is a common vascular amyloidogenic peptide recently implicated in Alzheimer's disease (AD) and vascular dementia and its pathology remains unknown. We aim to identify changes in transcriptomic profiles and pathways in human brain microvascular endothelial cells (HBMVECs) exposed to medin, compare that to exposure to ß-amyloid (Aß) and evaluate protection by monosialoganglioside-containing nanoliposomes (NL). HBMVECs were exposed for 20 h to medin (5 µM) without or with Aß(1-42) (2 µM) or NL (300 µg/mL), and RNA-seq with signaling pathway analyses were performed. Separately, reverse transcription polymerase chain reaction of select identified genes was done in HBMVECs treated with medin (5 µM) without or with NFκB inhibitor RO106-9920 (10 µM) or NL (300 µg/mL). Medin caused upregulation of pro-inflammatory genes that was not aggravated by Aß42 co-treatment but reversed by NL. Pathway analysis on differentially expressed genes revealed multiple pro-inflammatory signaling pathways, such as the tumor necrosis factor (TNF) and the nuclear factor-κB (NFkB) signaling pathways, were affected specifically by medin treatment. RO106-9920 and NL reduced medin-induced pro-inflammatory activation. Medin induced endothelial cell pro-inflammatory signaling in part via NFκB that was reversed by NL. This could have potential implications in the pathogenesis and treatment of vascular aging, AD and vascular dementia.


Subject(s)
Alzheimer Disease , Dementia, Vascular , Humans , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Dementia, Vascular/metabolism , Endothelial Cells/metabolism , Transcriptome
2.
J Neurotrauma ; 39(19-20): 1429-1441, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35593008

ABSTRACT

Severe traumatic brain injury (TBI) results in cognitive dysfunction in part due to vascular perturbations. In contrast, the long-term vasculo-cognitive pathophysiology of mild TBI (mTBI) remains unknown. We evaluated mTBI effects on chronic cognitive and cerebrovascular function and assessed their interrelationships. Sprague-Dawley rats received midline fluid percussion injury (n = 20) or sham (n = 21). Cognitive function was assessed (3- and 6-month novel object recognition [NOR], novel object location [NOL], and temporal order object recognition [TOR]). Six-month cerebral blood flow (CBF) and cerebral blood volume (CBV) using contrast magnetic resonance imaging (MRI) and ex vivo circle of Willis artery endothelial and smooth muscle-dependent function were measured. mTBI rats showed significantly impaired NOR, with similar trends (non-significant) in NOL/TOR. Regional CBF and CBV were similar in sham and mTBI. NOR correlated with CBF in lateral hippocampus, medial hippocampus, and primary somatosensory barrel cortex, whereas it inversely correlated with arterial smooth muscle-dependent dilation. Six-month baseline endothelial and smooth muscle-dependent arterial function were similar among mTBI and sham, but post-angiotensin 2 stimulation, mTBI showed no change in smooth muscle-dependent dilation from baseline response, unlike the reduction in sham. mTBI led to chronic cognitive dysfunction and altered angiotensin 2-stimulated smooth muscle-dependent vasoreactivity. The findings of persistent pathophysiological consequences of mTBI in this animal model add to the broader understanding of chronic pathophysiological sequelae in human mild TBI.


Subject(s)
Brain Concussion , Cerebrovascular Circulation , Cognition , Animals , Humans , Rats , Angiotensins , Brain Concussion/complications , Brain Concussion/pathology , Rats, Sprague-Dawley
3.
Stroke ; 53(2): e37-e41, 2022 02.
Article in English | MEDLINE | ID: mdl-34743535

ABSTRACT

BACKGROUND AND PURPOSE: Neuroprotective strategies for stroke remain inadequate. Nanoliposomes comprised of phosphatidylcholine, cholesterol, and monosialogangliosides (nanoliposomes) induced an antioxidant protective response in endothelial cells exposed to amyloid insults. We tested the hypotheses that nanoliposomes will preserve human neuroblastoma (SH-SY5Y) and human brain microvascular endothelial cells viability following oxygen-glucose deprivation (OGD)-reoxygenation and will reduce injury in mice following middle cerebral artery occlusion. METHODS: SH-SY5Y and human brain microvascular endothelial cells were exposed to oxygen-glucose deprivation-reoxygenation (3 hours 0.5%-1% oxygen and glucose-free media followed by 20-hour ambient air/regular media) without or with nanoliposomes (300 µg/mL). Viability was measured (calcein-acetoxymethyl fluorescence) and protein expression of antioxidant proteins HO-1 (heme oxygenase-1), NQO1 (NAD[P]H quinone dehydrogenase 1), and SOD1 (superoxide dismutase 1) were measured by Western blot. C57BL/6J mice were treated with saline (n=8) or nanoliposomes (10 mg/mL lipid, 200 µL, n=7) while undergoing 60-minute middle cerebral artery occlusion followed by reperfusion. Day 2 postinjury neurological impairment score and infarction size were compared. RESULTS: SH-SY5Y and human brain microvascular endothelial cells showed reduced viability post-oxygen-glucose deprivation-reoxygenation that was reversed by nanoliposomes. Nanoliposomes increased protein expressions of HO-1, NQO1 in both cell types and SOD1 in human brain microvascular endothelial cells. Nanoliposomes-treated mice showed reduced neurological impairment and brain infarct size (18.8±2% versus 27.3±2.3%, P=0.017) versus controls. CONCLUSIONS: Nanoliposomes reduced stroke injury in mice subjected to middle cerebral artery occlusion likely through induction of an antioxidant protective response. Nanoliposome is a candidate novel agent for stroke.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Liposomes/therapeutic use , Nanoparticles/therapeutic use , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Animals , Antioxidants/metabolism , Cell Line , Endothelium, Vascular/pathology , Glucose/deficiency , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Humans , Hypoxia , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Microvessels/pathology , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , NAD(P)H Dehydrogenase (Quinone)/genetics , Reperfusion Injury/pathology , Stroke/etiology , Stroke/pathology , Superoxide Dismutase-1/biosynthesis , Superoxide Dismutase-1/genetics
4.
Alzheimers Dement (Amst) ; 12(1): e12072, 2020.
Article in English | MEDLINE | ID: mdl-32875054

ABSTRACT

INTRODUCTION: Medin, an aging-associated amyloidogenic protein, induces cerebrovascular dysfunction and inflammation. We investigated the relationship between cerebrovascular medin and Alzheimer's disease (AD) and vascular dementia (VaD). METHODS: Cerebral arteriole medin was quantified from 91 brain donors with no dementia (ND), AD, VaD, or combined AD and VaD. Correlation analyses evaluated the relationship between arteriole medin, and plaques, tangles, or white matter lesions (WML). Receiver operating characteristic and regression analyses assessed whether medin is predictive of AD or VaD versus other cerebrovascular pathologies (circle of Willis [CoW] atherosclerosis and cerebral amyloid angiopathy [CAA]). RESULTS: Arteriole medin was higher in those with AD, VaD, or combined AD/VaD versus ND (P < .05), and correlated with tangle, plaque, and WML, but not CAA or CoW atherosclerosis. Among cerebrovascular pathologies, medin was the strongest predictor of AD diagnosis, whereas CoW atherosclerosis and arteriole medin were predictors of VaD. DISCUSSION: Cerebral arteriole medin is associated with and could be a potential novel risk factor or biomarker for AD and VaD.

5.
J Am Heart Assoc ; 9(2): e014810, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31928157

ABSTRACT

Background The function of medin, one of the most common human amyloid proteins that accumulates in the vasculature with aging, remains unknown. We aim to probe medin's role in cerebrovascular disease by comparing cerebral arterial medin content between cognitively normal and vascular dementia (VaD) patients and studying its effects on endothelial cell (EC) immune activation and neuroinflammation. We also tested whether monosialoganglioside-containing nanoliposomes could reverse medin's adverse effects. Methods and Results Cerebral artery medin and astrocyte activation were measured and compared between VaD and cognitively normal elderly brain donors. ECs were exposed to physiologic dose of medin (5 µmol/L), and viability and immune activation (interleukin-8, interleukin-6, intercellular adhesion molecule-1, and plasminogen activator inhibitor-1) were measured without or with monosialoganglioside-containing nanoliposomes (300 µg/mL). Astrocytes were exposed to vehicle, medin, medin-treated ECs, or their conditioned media, and interleukin-8 production was compared. Cerebral collateral arterial and parenchymal arteriole medin, white matter lesion scores, and astrocyte activation were higher in VaD versus cognitively normal donors. Medin induced EC immune activation (increased interleukin-8, interleukin-6, intercellular adhesion molecule-1, and plasminogen activator inhibitor-1) and reduced EC viability, which were reversed by monosialoganglioside-containing nanoliposomes. Interleukin-8 production was augmented when astrocytes were exposed to medin-treated ECs or their conditioned media. Conclusions Cerebral arterial medin is higher in VaD compared with cognitively normal patients. Medin induces EC immune activation that modulates astrocyte activation, and its effects are reversed by monosialoganglioside-containing nanoliposomes. Medin is a candidate novel risk factor for aging-related cerebrovascular disease and VaD.


Subject(s)
Antigens, Surface/toxicity , Astrocytes/drug effects , Cell Communication/drug effects , Cerebral Arteries/drug effects , Dementia, Vascular/drug therapy , Endothelial Cells/drug effects , Gangliosides/pharmacology , Milk Proteins/toxicity , Nanoparticles , Aged , Aged, 80 and over , Astrocytes/immunology , Astrocytes/metabolism , Astrocytes/pathology , Case-Control Studies , Cell Survival/drug effects , Cells, Cultured , Cerebral Arteries/immunology , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Coculture Techniques , Dementia, Vascular/immunology , Dementia, Vascular/metabolism , Dementia, Vascular/pathology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Liposomes , Male , Oxidative Stress/drug effects , Signal Transduction
6.
Am J Physiol Heart Circ Physiol ; 315(2): H284-H290, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29775413

ABSTRACT

Clinical and preclinical studies have suggested a link between cardiovascular disease and dementia disorders, but the role of the collateral brain circulation in cognitive dysfunction remains unknown. We aimed to test the hypothesis that leptomeningeal arteriole (LMA) function and response to metabolic stressors differ among subjects with dementia, mild cognitive impairment (MCI), and normal cognition (CN). After rapid autopsy, LMAs were isolated from subjects with CN ( n = 10), MCI ( n = 12), or dementia [ n = 42, Alzheimer's disease (AD), vascular dementia (VaD), or other dementia], and endothelial and smooth muscle-dependent function were measured at baseline and after exposure to ß-amyloid (2 µM), palmitic acid (150 µM), or medin (5 µM) and compared. There were no differences among the groups in baseline endothelial function (maximum dilation to acetylcholine, CN: 74.1 ± 9.7%, MCI: 67.1 ± 4.8%, AD: 74.7 ± 2.8%, VaD: 72.0 ± 5.3%, and other dementia: 68.0 ± 8.0%) and smooth muscle-dependent function (CN: 93.4 ± 3.0%, MCI: 83.3 ± 4.1%, AD: 91.8 ± 1.7%, VaD: 91.7 ± 2.4%, and other dementia: 87.9 ± 4.9%). There was no correlation between last cognitive function score and baseline endothelial or smooth muscle-dependent function. LMA endothelial function and, to a lesser extent, smooth muscle-dependent function were impaired posttreatment with ß-amyloid, palmitic acid, and medin. Posttreatment LMA responses were not different between subjects with CN/MCI vs. dementia. Baseline responses and impaired vasoreactivity after treatment with metabolic stressors did not differ among subjects with CN, MCI, and dementia. The results suggest that the cognitive dysfunction in dementia disorders is not attributable to differences in baseline brain collateral circulation function but may be influenced by exposure of the vasculature to metabolic stressors.


Subject(s)
Brain/blood supply , Cognitive Dysfunction/physiopathology , Dementia/physiopathology , Aged, 80 and over , Arterioles/physiopathology , Brain/physiopathology , Endothelium, Vascular/physiopathology , Female , Hemodynamics , Humans , Male , Myocytes, Smooth Muscle/physiology
7.
J Liposome Res ; 28(2): 97-105, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28103719

ABSTRACT

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.


Subject(s)
Amyloidogenic Proteins/metabolism , Amyloidosis/drug therapy , Clusterin/administration & dosage , Endothelium, Vascular/drug effects , Liposomes/chemistry , Nanoparticles/chemistry , Acetylcholine/chemistry , Arterioles/drug effects , Arterioles/metabolism , Cholesterol/chemistry , Clusterin/chemistry , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Humans , Papaverine/chemistry , Particle Size , Phosphatidylcholines/chemistry , Polyethylene Glycols/chemistry , Vasodilation/drug effects
8.
Cardiovasc Res ; 113(11): 1389-1402, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28859297

ABSTRACT

AIMS: Medin is a common amyloidogenic protein in humans that accumulates in arteries with advanced age and has been implicated in vascular degeneration. Medin's effect on endothelial function remains unknown. The aims are to assess medin's effects on human arteriole endothelial function and identify potential mechanisms underlying medin-induced vascular injury. METHODS AND RESULTS: Ex vivo human adipose and leptomeningeal arterioles were exposed (1 h) to medin (0.1, 1, or 5 µM) without or with FPS-ZM1 [100 µM, receptor for advanced glycation endproducts (RAGE)-specific inhibitor] and endothelium-dependent function (acetylcholine dilator response) and endothelium-independent function (dilator response to nitric oxide donor diethylenetriamine NONOate) were compared with baseline control. Human umbilical vein endothelial cells were exposed to medin without or with FPS-ZM1 and oxidative and nitrative stress, cell viability, and pro-inflammatory signaling measures were obtained. Medin caused impaired endothelial function (vs. baseline response: -45.2 ± 5.1 and -35.8 ± 7.9% in adipose and leptomeningeal arterioles, respectively, each P < 0.05). Dilator response to NONOate was not significantly changed. Medin decreased arteriole and endothelial cell nitric oxide production, increased superoxide production, reduced endothelial cell viability, proliferation, and migration. Medin increased gene and protein expression of interleukin-6 and interleukin-8 via activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB). Medin-induced endothelial dysfunction and oxidative stress were reversed by antioxidant polyethylene glycol superoxide dismutase and by RAGE inhibitor FPS-ZM1. CONCLUSIONS: Medin causes human microvascular endothelial dysfunction through oxidative and nitrative stress and promotes pro-inflammatory signaling in endothelial cells. These effects appear to be mediated via RAGE. The findings represent a potential novel mechanism of vascular injury.


Subject(s)
Endothelium, Vascular/metabolism , Receptor for Advanced Glycation End Products/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antioxidants/pharmacology , Arterioles/metabolism , Benzamides/pharmacology , Endothelium, Vascular/drug effects , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Male , Middle Aged , Oxidative Stress/drug effects , Superoxides/metabolism , Vasodilator Agents/pharmacology
9.
J Am Heart Assoc ; 5(6)2016 06 13.
Article in English | MEDLINE | ID: mdl-27412900

ABSTRACT

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.


Subject(s)
Cholesterol/administration & dosage , Endothelium, Vascular/drug effects , Gangliosides/administration & dosage , Immunoglobulin Light-chain Amyloidosis/complications , Phosphatidylcholines/administration & dosage , Vascular Diseases/prevention & control , Adipose Tissue/blood supply , Arterioles/drug effects , Arterioles/physiology , Cell Survival/physiology , Drug Combinations , Endothelial Cells/metabolism , Gene Knockdown Techniques/methods , Human Umbilical Vein Endothelial Cells , Humans , Immunoglobulin Light-chain Amyloidosis/prevention & control , Male , Middle Aged , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/genetics , Nanoparticles/administration & dosage , Nitric Oxide/physiology , Nitric Oxide Synthase Type III/metabolism , Papaverine/pharmacology , Peroxynitrous Acid/biosynthesis , RNA Interference/physiology , RNA, Small Interfering/physiology , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Transfection , Vascular Diseases/physiopathology , Vasodilator Agents/pharmacology
10.
J Cereb Blood Flow Metab ; 36(2): 405-12, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26661197

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides , Arterioles/pathology , Endothelium, Vascular/pathology , Liposomes/therapeutic use , Peptide Fragments , Vascular Diseases/chemically induced , Vascular Diseases/prevention & control , Acetylcholine/antagonists & inhibitors , Acetylcholine/pharmacology , Adipose Tissue/blood supply , Cholesterol/administration & dosage , Cholesterol/therapeutic use , Humans , In Vitro Techniques , Male , Meninges/blood supply , Middle Aged , NG-Nitroarginine Methyl Ester/pharmacology , Nanoparticles/therapeutic use , Nitric Oxide/biosynthesis , Phosphatidic Acids/administration & dosage , Phosphatidic Acids/therapeutic use , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/therapeutic use , Vascular Diseases/pathology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
11.
Biophys J ; 109(11): 2363-70, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26636947

ABSTRACT

The aggregation and fibril deposition of amyloid proteins have been implicated in a range of neurodegenerative and vascular diseases, and yet the underlying molecular mechanisms are poorly understood. Here, we use a combination of cell-based assays, biophysical analysis, and atomic force microscopy to investigate the potential involvement of oxidative stress in aortic medial amyloid (AMA) pathogenesis and deposition. We show that medin, the main constituent of AMA, can induce an environment rich in oxidative species, increasing superoxide and reducing bioavailable nitric oxide in human cells. We investigate the role that this oxidative environment may play in altering the aggregation process of medin and identify potential posttranslational modification sites where site-specific modification and interaction can be unambiguously demonstrated. In an oxidizing environment, medin is nitrated at tyrosine and tryptophan residues, with resultant effects on morphology that lead to longer fibrils with increased toxicity. This provides further motivation to investigate the role of oxidative stress in AMA pathogenicity.


Subject(s)
Antigens, Surface/toxicity , Aorta/metabolism , Milk Proteins/toxicity , Oxidative Stress/drug effects , Antigens, Surface/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Milk Proteins/metabolism , Nitrates/metabolism
12.
Diabetes ; 64(7): 2624-35, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25720388

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Endothelial Cells/drug effects , Peptides/pharmacology , Receptors, Glucagon/agonists , Vasodilation/drug effects , Venoms/pharmacology , AMP-Activated Protein Kinases/physiology , Blood Glucose/analysis , Cells, Cultured , Cross-Over Studies , Diabetes Mellitus, Type 2/physiopathology , Double-Blind Method , Endothelial Cells/physiology , Exenatide , Female , Glucagon-Like Peptide-1 Receptor , Humans , Male , Receptors, Glucagon/physiology , Triglycerides/blood
13.
J Neurosci Methods ; 235: 123-9, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25004204

ABSTRACT

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.


Subject(s)
Adipose Tissue/blood supply , Amyloid beta-Peptides , Arterioles/physiopathology , Endothelial Cells/physiology , Meninges/blood supply , Peptide Fragments , Abdomen/blood supply , Acetylcholine/pharmacology , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Female , Humans , Male , Middle Aged , Oxidative Stress/physiology , Papaverine/pharmacology , Reactive Oxygen Species/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vasodilator Agents/pharmacology
14.
J Liposome Res ; 24(1): 69-73, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24236475

ABSTRACT

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.


Subject(s)
Amyloid/chemistry , Amyloidosis/drug therapy , Endothelium/drug effects , Liposomes/therapeutic use , Nanoparticles/therapeutic use , Aged , Apoptosis/drug effects , Endothelium/injuries , Endothelium/pathology , Heart Failure/drug therapy , Heart Failure/pathology , Humans , Liposomes/chemistry , Male , Middle Aged , Nanoparticles/chemistry , Proteostasis Deficiencies/drug therapy
15.
Atherosclerosis ; 225(1): 220-3, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22981431

ABSTRACT

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.


Subject(s)
Amyloidosis/physiopathology , Clusterin/physiology , Endothelial Cells/physiology , Immunoglobulin Light Chains , Arterioles/physiopathology , Cell Death/drug effects , Clusterin/biosynthesis , DNA Fragmentation/drug effects , Endothelial Cells/drug effects , Female , Humans , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light Chains/pharmacology , Male , Middle Aged , Vasodilation/drug effects
16.
Am J Physiol Heart Circ Physiol ; 301(6): H2305-12, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21963839

ABSTRACT

Light chain amyloidosis (AL) involves overproduction of amyloidogenic light chain proteins (LC) leading to heart failure, yet the mechanisms underlying tissue toxicity remain unknown. We hypothesized that LC induces endothelial dysfunction in non-AL human microvasculature and apoptotic injury in human coronary artery endothelial cells (HCAECs). Adipose arterioles (n = 34, 50 ± 3 yr) and atrial coronary arterioles (n = 19, 68 ± 2 yr) from non-AL subjects were cannulated. Adipose arteriole dilator responses to acetylcholine/papaverine were measured at baseline and 1 h exposure to LC (20 µg/ml) from biopsy-proven AL subjects (57 ± 11 yr) without and with antioxidant cotreatment. Coronary arteriole dilation to bradykinin/papaverine was measured post-LC exposure. HCAECs were exposed to 1 or 24 h of LC. LC reduced dilation to acetylcholine (10(-4) M: 41.6 ± 7 vs. 85.8 ± 2.2% control, P < 0.001) and papaverine (81.4 ± 4.6 vs. 94.8 ± 1.3% control, P < 0.01) in adipose arterioles and to bradykinin (10(-6) M: 68.6 ± 6.2 vs. 90.9 ± 1.6% control, P < 0.001) but not papaverine in coronary arterioles. There was an increase in superoxide and peroxynitrite in arterioles treated with LC. Adipose arteriole dilation was restored by cotreatment with polyethylene glycol-superoxide dismutase and tetrahydrobiopterin but only partially restored by mitoquinone (mitochondria-targeted antioxidant) and gp91ds-tat (NADPH oxidase inhibitor). HCAECs exposed to LC showed reduced NO and increased superoxide, peroxynitrite, annexin-V, and propidium iodide compared with control. Brief exposure to physiological amounts of LC induced endothelial dysfunction in human adipose and coronary arterioles and increased apoptotic injury in coronary artery endothelial cells likely as a result of oxidative stress, reduced NO bioavailability, and peroxynitrite production. Microvascular dysfunction and injury is a novel mechanism underlying AL pathobiology and is a potential target for therapy.


Subject(s)
Adipose Tissue/blood supply , Amyloidosis/metabolism , Apoptosis , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Immunoglobulin Light Chains/metabolism , Vasodilation , Aged , Amyloidosis/pathology , Amyloidosis/physiopathology , Antioxidants/pharmacology , Arterioles/metabolism , Arterioles/pathology , Arterioles/physiopathology , Case-Control Studies , Cells, Cultured , Coronary Vessels/drug effects , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Dose-Response Relationship, Drug , Endothelial Cells/pathology , Female , Humans , Male , Middle Aged , Nitric Oxide/metabolism , Oxidative Stress , Peroxynitrous Acid/metabolism , Superoxides/metabolism , Time Factors , Vasodilation/drug effects , Vasodilator Agents/pharmacology
17.
Int J Cardiol ; 145(1): 67-8, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-19446898

ABSTRACT

Light chain amyloidosis (AL) is a plasma cell dyscrasia associated with production of amyloidogenic immunoglobulin light chains (LC). Despite its often fatal course, the mechanism of injury remains unknown. We tested the hypothesis that AL is associated with oxidative stress by comparing serum protein carbonyl (a marker of protein oxidation and oxidative stress) in AL subjects (n=23, 60 ± 11 years) vs. controls (n=9, 54 ± 2 years); we also measured superoxide production (n=11) and dilator response to sodium nitroprusside (SNP, n=6) in isolated non-AL human adipose arterioles exposed to LC (20 µg/mL) purified from AL subjects for 1 h vs. control. Protein carbonyl was higher in AL patients (0.19 ± 0.04 vs. 0.003 ± 0.003 nmol/mg control, p=0.002). Post-exposure to LC proteins, arteriole superoxide was higher (1.89 ± 0.36 times control, p=0.03) with impaired dilation to SNP (10(-4) M, 54 ± 6 vs. 86 ± 4%, p=0.01, logEC50 -3.7 ± 0.2 vs. -6.7 ± 0.6, p=0.002). AL is associated with systemic oxidative stress and brief acute exposure to AL light chain proteins induces oxidative stress and microvascular dysfunction in human adipose arterioles. This novel mechanism of injury may be important in AL pathophysiology.


Subject(s)
Amyloidosis/immunology , Amyloidosis/metabolism , Arterioles/immunology , Immunoglobulin Light Chains/adverse effects , Microvessels/immunology , Oxidative Stress/immunology , Aged , Amyloidosis/physiopathology , Arterioles/metabolism , Biomarkers/blood , Female , Humans , Immunoglobulin Light Chains/biosynthesis , Immunoglobulin Light Chains/blood , Male , Microvessels/metabolism , Middle Aged , Protein Carbonylation/immunology
18.
BMC Med Phys ; 9: 5, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19416541

ABSTRACT

BACKGROUND: Light chain amyloidosis (AL) is a rare plasma cell dyscrasia associated with poor survival especially in the setting of heart failure. Late gadolinium enhancement (LGE) on cardiac MRI was recently found to correlate with myocardial amyloid deposition but the prognostic role is not established. The aim is to determine the prognostic significance of LGE in AL by comparing long term survival of AL patients with and without LGE. METHODS: Twenty nine consecutive patients (14 females; 62 +/- 11 years) with biopsy-proven AL undergoing cardiac MRI with gadolinium as part of AL workup were included. Survival was prospectively followed 29 months (median) following MRI and compared between those with and without LGE by Kaplan-Meier and log-rank analyses. RESULTS: LGE was positive in 23 subjects (79%) and negative in 6 (21%). Left ventricular ejection fraction was 66 +/- 17% in LGE-positive and 69 +/- 12% in LGE-negative patients (p = 0.8). Overall 1-year mortality was 36%. On follow-up, 14/23 LGE-positive and none of LGE-negative patients died (log rank p = 0.0061). Presenting New York Heart Association heart failure class was also associated with poor survival (p = 0.0059). Survival between two LGE groups stratified by heart failure class still showed a significant difference by a stratified log-rank test (p = 0.04). CONCLUSION: Late gadolinium enhancement is common and is associated with poor long-term survival in light chain amyloidosis, even after adjustment for heart failure class presentation. The prognostic significance of late gadolinium enhancement in this disease may be useful in patient risk-stratification.

19.
Cardiovasc Ultrasound ; 6: 40, 2008 Aug 07.
Article in English | MEDLINE | ID: mdl-18687125

ABSTRACT

BACKGROUND: Light chain amyloidosis (AL) is a rare but often fatal disease due to intractable heart failure. Amyloid deposition leads to diastolic dysfunction and often preserved ejection fraction. We hypothesize that AL is associated with regional systolic dyssynchrony. The aim is to compare left ventricular (LV) regional synchrony in AL subjects versus healthy controls using 16-segment dyssynchrony index measured from 3-dimension-al (3D) echocardiography. METHODS: Cardiac 3D echocardiography full volumes were acquired in 10 biopsy-proven AL subjects (60 +/- 3 years, 5 females) and 10 healthy controls (52 +/- 1 years, 5 females). The LV was subdivided into 16 segments and the time from end-diastole to the minimal systolic volume for each of the 16 segments was expressed as a percent of the cycle length. The standard deviations of these times provided a 16-segment dyssynchrony index (16-SD%). 16-SD% was compared between healthy and AL subjects. RESULTS: Left ventricular ejection fraction was comparable (control vs. AL: 62.4 +/- 0.6 vs. 58.6 +/- 2.8%, p = NS). 16-SD% was significantly higher in AL versus healthy subjects (5.93 +/- 4.4 vs. 1.67 +/- 0.87%, p = 0.003). 16-SD% correlated with left ventricular mass index (R 0.45, p = 0.04) but not to left ventricular ejection fraction. CONCLUSION: Light chain amyloidosis is associated with left ventricular regional systolic dyssynchrony. Regional dyssynchrony may be an unrecognized mechanism of heart failure in AL subjects.


Subject(s)
Amyloidosis/diagnostic imaging , Cardiomyopathies/diagnostic imaging , Echocardiography, Three-Dimensional , Ventricular Dysfunction, Left/diagnostic imaging , Amyloidosis/complications , Amyloidosis/physiopathology , Cardiomyopathies/physiopathology , Female , Humans , Male , Middle Aged , Stroke Volume , Systole , Ventricular Dysfunction, Left/complications , Ventricular Dysfunction, Left/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...