Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Aging Cell ; 19(8): e13196, 2020 08.
Article in English | MEDLINE | ID: mdl-32691484

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive disease thought to result from impaired lung repair following injury and is strongly associated with aging. While vascular alterations have been associated with IPF previously, the contribution of lung vasculature during injury resolution and fibrosis is not well understood. To compare the role of endothelial cells (ECs) in resolving and non-resolving models of lung fibrosis, we applied bleomycin intratracheally to young and aged mice. We found that injury in aged mice elicited capillary rarefaction, while injury in young mice resulted in increased capillary density. ECs from the lungs of injured aged mice relative to young mice demonstrated elevated pro-fibrotic and reduced vascular homeostasis gene expression. Among the latter, Nos3 (encoding the enzyme endothelial nitric oxide synthase, eNOS) was transiently upregulated in lung ECs from young but not aged mice following injury. Young mice deficient in eNOS recapitulated the non-resolving lung fibrosis observed in aged animals following injury, suggesting that eNOS directly participates in lung fibrosis resolution. Activation of the NO receptor soluble guanylate cyclase in human lung fibroblasts reduced TGFß-induced pro-fibrotic gene and protein expression. Additionally, loss of eNOS in human lung ECs reduced the suppression of TGFß-induced lung fibroblast activation in 2D and 3D co-cultures. Altogether, our results demonstrate that persistent lung fibrosis in aged mice is accompanied by capillary rarefaction, loss of EC identity, and impaired eNOS expression. Targeting vascular function may thus be critical to promote lung repair and fibrosis resolution in aging and IPF.


Subject(s)
Bleomycin/adverse effects , Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/chemically induced , Lung/pathology , Animals , Humans , Mice
2.
J Cereb Blood Flow Metab ; 40(2): 392-403, 2020 02.
Article in English | MEDLINE | ID: mdl-30614363

ABSTRACT

Cerebral amyloid angiopathy (CAA) is present in over half of the elderly population and in 80-90% of Alzheimer's disease (AD) patients. CAA is defined by the deposition of beta amyloid (Aß) in small cerebral arteries and capillaries. Cardiovascular risk factors are associated with an increased incidence of CAA. We utilized 18-month-old endothelial nitric oxide synthase (eNOS) heterozygous knockout (+/-) mice, a clinically relevant model of endothelial dysfunction, to examine the role of endothelial nitric oxide (NO) in vascular Aß accumulation. eNOS+/- mice had significantly higher vascular levels of Aß40 (P < 0.05). Aß42 was not detected. There was no difference in Aß in brain tissue. Amyloid precursor protein and ß-site APP cleavage enzyme 1 protein levels were unaltered, while levels of the α-secretase enzyme, a disintegrin and metalloproteinase 10, were significantly lower in eNOS + /- microvascular tissue (P < 0.05). Insulin degrading enzyme and low-density lipoprotein receptor-related protein 1 were significantly increased in eNOS+/- microvascular tissue, most likely an adaptive response to locally higher Aß concentrations. Lastly, catalase and CuZn superoxide dismutase were significantly elevated in eNOS+/- microvascular tissue (P < 0.05). These data demonstrate decreased availability of endothelial NO leads to increased cerebrovascular concentration of Aß along with compensatory mechanisms to protect the vasculature.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/metabolism , Brain , Cerebral Amyloid Angiopathy , Endothelium, Vascular , Nitric Oxide Synthase Type III/deficiency , Peptide Fragments/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Brain/blood supply , Brain/metabolism , Brain/pathology , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Peptide Fragments/genetics
3.
Circ Res ; 119(10): 1128-1134, 2016 Oct 28.
Article in English | MEDLINE | ID: mdl-27601478

ABSTRACT

RATIONALE: Alzheimer's disease has an unknown pathogenesis; however, cardiovascular risk factors are associated with a higher incidence of Alzheimer's disease. A defining feature of endothelial dysfunction induced by cardiovascular risk factors is reduced bioavailable endothelial nitric oxide (NO). We previously demonstrated that endothelial NO acts as an important signaling molecule in neuronal tissue. OBJECTIVE: We sought to determine the relationship between the loss of endothelial NO synthase (eNOS) and tau phosphorylation in neuronal tissue. METHODS AND RESULTS: We used eNOS knockout (-/-) mice as well as an Alzheimer's disease mouse model, amyloid precursor protein (APP)/PSEN1dE9+/- (PS1) that lacked eNOS (APP/PS1/eNOS-/-) to examine expression of tau kinases and tau phosphorylation. Brain tissue from eNOS-/- mice had statistically higher ratios of p25/p35, indicative of increased cyclin-dependent kinase 5 activity as compared with wild-type (n=8, P<0.05). However, tau phosphorylation was unchanged in eNOS-/- mice (P>0.05). Next, we determined the role of NO in tau pathology in APP/PS1/eNOS-/-. These mice had significantly higher levels of p25, a higher p25/p35 ratio (n=12-14; P<0.05), and significantly higher cyclin-dependent kinase 5 activity (n=4; P<0.001). Importantly, APP/PS1/eNOS-/- mice also had significantly increased tau phosphorylation (n=4-6; P<0.05). No other changes in amyloid pathology, antioxidant pathways, or neuroinflammation were observed in APP/PS1/eNOS-/- mice as compared with APP/PS1 mice. CONCLUSIONS: Our data suggests that loss of endothelial NO plays an important role in the generation of p25 and resulting tau phosphorylation in neuronal tissue. These findings provide important new insights into the molecular mechanisms linking endothelial dysfunction with the pathogenesis of Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Nitric Oxide Synthase Type III/deficiency , Phosphotransferases/biosynthesis , tau Proteins/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Antioxidants/metabolism , Brain Chemistry , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type III/physiology , Phosphorylation , Phosphotransferases/genetics , Presenilin-1/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins I/metabolism , Protein Processing, Post-Translational , Rats
4.
Circ J ; 80(7): 1499-503, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27238834

ABSTRACT

In the central nervous system endothelial nitric oxide (NO) is an essential molecule responsible for the preservation of the functional integrity of the neurovascular unit. NO causes vasodilatation and is an important inhibitor of platelet aggregation, smooth muscle cell proliferation, and white blood cell adhesion. In addition, endothelium-derived NO exerts anti-inflammatory and pro-angiogenic effects. More recently, it has been recognized that endothelial NO modulates the expression and processing of amyloid precursor protein in cerebrovascular endothelium and neuronal tissue. Studies in endothelial NO synthase (eNOS) knockout mice indicate that endothelial NO functions as a neurovascular protective molecule during aging. Indeed, genetic inactivation of eNOS exacerbates the detrimental effects of aging on cerebrovascular, microglial, and neuronal functions as well as on cognition. These findings suggest that the preservation of healthy endothelium and normal function of eNOS might be important therapeutic targets. Because the beneficial effects of NO are mostly mediated by the activation of guanylate cyclase/cyclic GMP signaling, inhibitors of phosphodiesterase isoforms, or activation of this signaling with exercise, may offer therapeutic opportunities in the prevention and treatment of aging-induced cognitive decline and Alzheimer's disease. Most recent advances in understanding the molecular mechanisms linking loss of endothelial NO with cognitive decline will be discussed in this review. (Circ J 2016; 80: 1499-1503).


Subject(s)
Alzheimer Disease/metabolism , Endothelium, Vascular/metabolism , Microglia/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Second Messenger Systems , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Animals , Cognition , Cyclic GMP/metabolism , Humans , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism
5.
PLoS One ; 10(12): e0144062, 2015.
Article in English | MEDLINE | ID: mdl-26629821

ABSTRACT

The hippocampus is one of the earliest and most affected regions in Alzheimer's disease (AD), followed by the cortex while the cerebellum is largely spared. Importantly, endothelial dysfunction is a common feature of cerebral blood vessels in AD. In this study, we sought to determine if regional heterogeneity of cerebral microvessels might help explain the susceptibility of the hippocampus and cortex as compared to the cerebellum. We isolated microvessels from wild type mice from the cerebellum, cortex, and hippocampus to characterize their vascular phenotype. Superoxide anion was significantly higher in microvessels isolated from the cortex and hippocampus as compared to the cerebellum. Importantly, protein levels of NADPH oxidase (NOX)-2 and NOX-4 were significantly higher in the cortical and hippocampal microvessels as compared to microvessels from the cerebellum. In addition, expression of manganese superoxide dismutase protein was significantly lower in microvessels from the cortex and hippocampus as compared to cerebellum while other antioxidant enzymes were unchanged. There was no difference in eNOS protein expression between the microvessels of the three brain regions; however, bioavailability of tetrahydrobiopterin (BH4), an essential cofactor for eNOS activity, was significantly reduced in microvessels from the hippocampus and cortex as compared to the cerebellum. Higher levels of superoxide and reduced tetrahydrobiopterin bioavailability may help explain the vulnerability of the hippocampus and cortical microvessels to oxidative stress and development of endothelial dysfunction.


Subject(s)
Cerebellum/pathology , Cerebral Cortex/pathology , Hippocampus/pathology , Microvessels/pathology , Oxidative Stress/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Antioxidants/metabolism , Biological Availability , Biopterins/analogs & derivatives , Biopterins/metabolism , Cerebellum/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Microvessels/metabolism , Nitric Oxide Synthase Type III/metabolism , Superoxide Dismutase/metabolism , Superoxides/metabolism
6.
PLoS One ; 10(7): e0132604, 2015.
Article in English | MEDLINE | ID: mdl-26161952

ABSTRACT

Microgliosis is a major hallmark of Alzheimer's disease (AD) brain pathology. Aß peptide is hypothesized to act as a stimulus for microglia leading to activation of non-receptor tyrosine kinases and subsequent secretion of pro-inflammatory cytokines. Therefore, the signaling pathways mediating microglial activation may be important therapeutic targets of anti-inflammatory therapy for AD. Four novel compounds were chosen after high throughput screening kinase activity assays determined them as potential Lyn kinase inhibitors. Their kinase inhibitory and anti-inflammatory effect on Aß-stimulated activation was assessed using the murine microglial cell line, BV2. Cells were treated with the compounds to determine effects on active, phosphorylated levels of Src family kinases, Src and Lyn, as well as MAP kinases ERK, JNK and p38. Only one compound, LDDN-0003499, produced a dose dependent decrease in basal levels of active, phosphorylated Src and Lyn in the BV2 cells. LDDN-0003499 treatment also attenuated the Aß-stimulated increase in active, phosphorylated levels of Lyn/Src and TNFα and IL-6 secretion. This study identifies a novel small molecule Src family tyrosine kinase inhibitor with anti-inflammatory effects in response to Aß stimulation of microglia. Further in vitro/in vivo characterization of LDDN-0003499 as well as structural modification may provide a new tool for attenuating microglial-mediated brain inflammatory conditions such as that occurring in AD.


Subject(s)
Gliosis/pathology , src-Family Kinases/antagonists & inhibitors , Administration, Oral , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Caco-2 Cells , Cell Line , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Gliosis/enzymology , Humans , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Microsomes/drug effects , Microsomes/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Kinase Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/metabolism
7.
Eur Heart J ; 35(14): 888-94, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24357508

ABSTRACT

Endothelial nitric oxide (NO) is generated by constitutively active endothelial nitric oxide synthase (eNOS), an essential enzyme responsible for cardiovascular homeostasis. Historically, endothelial NO was first recognized as a major vasodilator involved in control of vasomotor function and local blood flow. In this review, our attention is focused on the emerging role of endothelial NO in linking cerebrovascular function with cognition. We will discuss the recognized ability of endothelial NO to modulate processing of amyloid precursor protein (APP), influence functional status of microglia, and affect cognitive function. Existing evidence suggests that the loss of NO in cultured human cerebrovascular endothelium causes increased expression of APP and ß-site APP-cleaving enzyme 1 (BACE1) thereby resulting in increased secretion of amyloid ß peptides (Aß1-40 and Aß1-42). Furthermore, increased expression of APP and BACE1 as well as increased production of Aß peptides was detected in the cerebral microvasculature and brain tissue of eNOS-deficient mice. Since Aß peptides are considered major cytotoxic molecules responsible for the pathogenesis of Alzheimer's disease, these observations support the concept that a loss of endothelial NO might significantly contribute to the initiation and progression of cognitive decline. In addition, genetic inactivation of eNOS causes activation of microglia and promotes a pro-inflammatory phenotype in the brain. Behavioural analysis revealed that eNOS-deficient mice exhibit impaired cognitive performance thereby indicating that selective loss of endothelial NO has a detrimental effect on the function of neuronal cells. Together with findings from prior studies demonstrating the ability of endothelial NO to affect synaptic plasticity, mitochondrial biogenesis, and function of neuronal progenitor cells, it is becoming apparent that the role of endothelial NO in the control of central nervous system function is very complex. We propose that endothelial NO represents the key molecule linking cerebrovascular and neuronal function.


Subject(s)
Alzheimer Disease/etiology , Nitric Oxide/physiology , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/blood supply , Cerebrovascular Circulation/physiology , Endothelium, Vascular/physiology , Humans , Mice , Microglia/physiology , Microvessels/metabolism , Neuronal Plasticity/physiology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/physiology , Oxidative Stress/physiology
8.
J Neurochem ; 127(5): 691-700, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23745722

ABSTRACT

Aging and the presence of cerebrovascular disease are associated with increased incidence of Alzheimer's disease. A common feature of aging and cerebrovascular disease is decreased endothelial nitric oxide (NO). We studied the effect of a loss of endothelium derived NO on amyloid precursor protein (APP) related phenotype in late middle aged (LMA) (14-15 month) endothelial nitric oxide synthase deficient (eNOS(-/-) ) mice. APP, ß-site APP cleaving enzyme (BACE) 1, and amyloid beta (Aß) levels were significantly higher in the brains of LMA eNOS(-/-) mice as compared with LMA wild-type controls. APP and Aß1-40 were increased in hippocampal tissue of eNOS(-/-) mice as compared with wild-type mice. LMA eNOS(-/-) mice displayed an increased inflammatory phenotype as compared with LMA wild-type mice. Importantly, LMA eNOS(-/-) mice performed worse in a radial arm maze test of spatial learning and memory as compared with LMA wild-type mice. These data suggest that chronic loss of endothelial NO may be an important contributor to both Aß related pathology and cognitive decline. Cardiovascular risk factors are associated with increased incidence of Alzheimer's disease (AD). A common feature of these risk factors is decreased endothelial nitric oxide (NO). We observed, in mice deficient in endothelial nitric oxide synthase, increased amyloid precursor protein (APP), ß-site APP cleaving enzyme 1, amyloid beta levels, microglial activation, and impaired spatial memory. This suggests chronic loss of endothelial NO may be an important contributor to the pathogenesis of sporadic AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Alzheimer Disease/epidemiology , Amyloid beta-Protein Precursor/metabolism , Animals , Encephalitis/epidemiology , Encephalitis/metabolism , Encephalitis/pathology , Hippocampus/blood supply , Hippocampus/enzymology , Hippocampus/pathology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Microvessels/enzymology , Microvessels/pathology , Nitric Oxide Synthase Type III/deficiency , Phenotype , Risk Factors
9.
J Alzheimers Dis ; 33(1): 29-33, 2013.
Article in English | MEDLINE | ID: mdl-22886025

ABSTRACT

Recently, we demonstrated in endothelial nitric oxide synthase deficient (eNOS-/-) mice that loss of endothelial NO led to increased protein levels of amyloid-ß protein precursor (AßPP), ß-site AßPP cleaving enzyme 1 (BACE1), and amyloid-ß (Aß) peptide. Therefore, our aim was to determine if NO supplementation in vivo would attenuate AßPP and BACE1 protein levels. cGMP levels were significantly increased while AßPP and BACE1 protein levels were statistically lower in cerebral microvessels from nitroglycerin-treated eNOS-/- mice as compared to vehicle-treated mice. Our findings support the concept that preservation of NO/cGMP signaling is an important modulator of expression and processing of AßPP.


Subject(s)
Amyloid Precursor Protein Secretases/biosynthesis , Amyloid beta-Protein Precursor/biosynthesis , Aspartic Acid Endopeptidases/biosynthesis , Cerebrovascular Circulation/physiology , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide/administration & dosage , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cerebrovascular Circulation/drug effects , Cyclic GMP/genetics , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcirculation/drug effects , Microcirculation/genetics , Nitric Oxide/genetics , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Nitroglycerin/pharmacology , Protein Processing, Post-Translational/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
10.
Neurochem Res ; 36(6): 994-1004, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21384098

ABSTRACT

Alpha (α)-synuclein neuronal effects are continually being defined although its role in regulating glial phenotypes remains unclear. An ability to regulate microglial activation was investigated using primary cultures from wild type and α-synuclein deficient mice (Snca-/-). Snca-/- microglia demonstrated increased secretion of the cytokine tumor necrosis factor-alpha (TNF-α), impaired phagocytic ability, elevated prostaglandin levels, and increased protein levels of key enzymes in lipid-mediated signaling events, cytosolic phospholipase (cPLA(2)), cyclooxygenase-2 (Cox-2) and phospholipase D2 (PLD2) when compared to wild type cells. Increased cytokine secretion and cPLA(2) and Cox-2 levels in Snca-/- microglia were partially attenuated by inhibiting PLD-dependent signaling with n-butanol treatment.


Subject(s)
Microglia/physiology , alpha-Synuclein/physiology , Animals , Blotting, Western , Mice , Mice, Knockout , alpha-Synuclein/genetics
11.
Circ Res ; 107(12): 1498-502, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21127294

ABSTRACT

RATIONALE: the exact etiology of sporadic Alzheimer disease (AD) is unclear, but it is interesting that several cardiovascular risk factors are associated with higher incidence of AD. The link between these risk factors and AD has yet to be identified; however, a common feature is endothelial dysfunction, specifically, decreased bioavailability of nitric oxide (NO). OBJECTIVE: to determine the relationship between endothelial derived NO and the expression and processing of amyloid precursor protein (APP). METHODS AND RESULTS: we used human brain microvascular endothelial cells to examine the role of NO in modulating APP expression and processing in vitro. Inhibition of endothelial nitric oxide synthase (eNOS) with the specific NOS inhibitor L-NAME (N(G)-nitro-l-arginine methyl ester) led to increased APP and BACE1 (ß-site APP-cleaving enzyme1) protein levels, as well as increased secretion of the amyloidogenic peptide amyloid ß (Aß) (control 10.93 ± 0.70 pg/mL; L-NAME 168.21 ± 27.38 pg/mL; P<0.001). To examine the role of NO in modulation of APP expression and processing in vivo, we used brain and cerebral microvessels from eNOS-deficient (eNOS(-/-)) mice. Brain tissue from eNOS(-/-) mice had statistically higher APP and BACE1 protein levels, as well as increased BACE1 enzyme activity and Aß (Aß(1)(-)(42) wild-type control, 0.737 pg/mg; eNOS(-/-), 1.475 pg/mg; P<0.05), compared with wild-type controls (n=6 to 8 animals per background). Brain microvessels from eNOS(-/-) mice also showed statistically higher BACE1 protein levels as compared with wild-type control. CONCLUSIONS: our data suggest that endothelial NO plays an important role in modulating APP expression and processing within the brain and cerebrovasculature. The NO/cGMP pathway may be an important therapeutic target in preventing and treating mild cognitive impairment, as well as AD.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , Brain/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/pathology , Nitric Oxide Synthase Type III/physiology , Alzheimer Disease , Amyloid Precursor Protein Secretases/analysis , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/analysis , Brain/blood supply , Brain/cytology , Brain Chemistry , Cerebrovascular Circulation , Cyclic GMP , Endothelial Cells/pathology , Endothelium, Vascular/physiopathology , Humans , Mice , Microcirculation , Nitric Oxide/physiology
12.
Neurobiol Aging ; 31(11): 1854-66, 2010 Nov.
Article in English | MEDLINE | ID: mdl-19058878

ABSTRACT

Amyloid precursor protein (APP) is a type 1 integral membrane protein, which is highly conserved and ubiquitously expressed. Numerous data suggest it functions in cellular adhesion. For example, APP binds components of the extracellular matrix to propagate intracellular signaling responses. In order to investigate adhesion-related changes in inflamed vasculature, brains from apolipoprotein E(-/-) (apoE(-/-)) mice were examined for changes related to APP then compared to human Alzheimer's disease (AD) brains. Cerebrovasculature from mouse apoE(-)/(-) and human AD brains revealed strong immunoreactivity for APP, APP phosphorylated at tyrosine residue 682 (pAPP) and Aß. Further, Western blot analyses from mouse apoE(-/-) and AD brains showed statistically higher protein levels of APP, pAPP and increased APP association with the tyrosine kinase, Src. Lastly, utilizing a modified Stamper-Woodruff adhesion assay, we demonstrated that adhesion of monocytic cells to apoE(-/-) and AD brain endothelium is partially APP dependent. These data suggest that endothelial APP function coupled with increased Aß production are involved in the vascular dysfunction associated with atherosclerosis and AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Apolipoproteins E/metabolism , Cell Adhesion/immunology , Endothelial Cells/metabolism , Monocytes/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/immunology , Animals , Apolipoproteins E/genetics , Brain/blood supply , Brain/immunology , Brain/metabolism , Brain/pathology , Endothelial Cells/immunology , Humans , Mice , Mice, Knockout , Monocytes/immunology , src-Family Kinases/metabolism
13.
J Neurosci ; 29(46): 14451-62, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19923279

ABSTRACT

Amyloid precursor protein (APP) is a ubiquitously expressed type 1 integral membrane protein. It has the ability to bind numerous extracellular matrix components and propagate signaling responses via its cytoplasmic phospho-tyrosine, (682)YENPTY(687), binding motif. We recently demonstrated increased protein levels of APP, phosphorylated APP (Tyr682), and beta-amyloid (Abeta) in brain vasculature of atherosclerotic and Alzheimer's disease (AD) tissue colocalizing primarily within the endothelial layer. This study demonstrates similar APP changes in peripheral vasculature from human and mouse apoE(-/-) aorta, suggesting that APP-related changes are not restricted to brain vasculature. Therefore, primary mouse aortic endothelial cells and human umbilical vein endothelial cells were used as a model system to examine the function of APP in endothelial cells. APP multimerization with an anti-N-terminal APP antibody, 22C11, to simulate ligand binding stimulated an Src kinase family-dependent increase in protein phospho-tyrosine levels, APP phosphorylation, and Abeta secretion. Furthermore, APP multimerization stimulated increased protein levels of the proinflammatory proteins, cyclooxygenase-2 and vascular cell adhesion molecule-1 also in an Src kinase family-dependent manner. Endothelial APP was also involved in mediating monocytic cell adhesion. Collectively, these data demonstrate that endothelial APP regulates immune cell adhesion and stimulates a tyrosine kinase-dependent response driving acquisition of a reactive endothelial phenotype. These APP-mediated events may serve as therapeutic targets for intervention in progressive vascular changes common to cerebrovascular disease and AD.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Endothelial Cells/enzymology , Protein-Tyrosine Kinases/metabolism , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/enzymology , Aorta, Abdominal/immunology , Cell Line, Tumor , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/immunology , Enzyme Activation/immunology , Humans , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
14.
J Neurosci ; 26(41): 10558-63, 2006 Oct 11.
Article in English | MEDLINE | ID: mdl-17035541

ABSTRACT

Recent Parkinson's disease research has focused on understanding the function of the cytosolic protein, alpha-synuclein, and its contribution to disease mechanisms. Within neurons, alpha-synuclein is hypothesized to have a role in regulating synaptic plasticity, vesicle release, and trafficking. In contrast, glial-expressed alpha-synuclein remains poorly described. Here, we examine the consequence of a loss of alpha-synuclein expression on microglial activation. Using a postnatal brain-derived culture system, we defined the phenotype of microglia from wild-type and knock-out alpha-synuclein mice (Scna-/-). Scna-/- microglia displayed a basally increased reactive phenotype compared with the wild-type cells and an exacerbated reactive phenotype after stimulation. They also exhibited dramatic morphologic differences compared with wild-type, presenting as large, ramified cells filled with vacuole-like structures. This corresponded with increased protein levels of activation markers, CD68 and beta1 integrin, in the Scna-/- cells. More importantly, Scna-/- microglia, after stimulation, secreted elevated levels of proinflammatory cytokines, TNFalpha (tumor necrosis factor alpha) and IL-6 (interleukin-6), compared with wild type. However, despite the reactive phenotype, Scna-/- cells had impaired phagocytic ability. We demonstrate for the first time that alpha-synuclein plays a critical role in modulating microglial activation state. We suggest that altered microglial alpha-synuclein expression will affect their phenotype as has already been demonstrated in neurons. This has direct ramifications for the contribution of microglia to the pathophysiology of disease, particularly in familial cases linked to altered alpha-synuclein expression.


Subject(s)
Gene Expression Regulation/physiology , Microglia/metabolism , Phenotype , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics , Animals , Cells, Cultured , Mice , Mice, Knockout , Microglia/cytology , alpha-Synuclein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...