Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Susan A. Austin is active.

Publication


Featured researches published by Susan A. Austin.


Circulation Research | 2010

Endothelial Nitric Oxide Modulates Expression and Processing of Amyloid Precursor Protein

Susan A. Austin; Anantha Vijay R. Santhanam; Zvonimir S. Katusic

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 (NG-nitro-L-arginine methyl ester) led to increased APP and BACE1 (&bgr;-site APP-cleaving enzyme1) protein levels, as well as increased secretion of the amyloidogenic peptide amyloid &bgr; (A&bgr;) (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 A&bgr; (A&bgr;1-42 wild-type control, 0.737pg/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.


European Heart Journal | 2014

Endothelial nitric oxide: protector of a healthy mind

Zvonimir S. Katusic; Susan A. Austin

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 β-site APP-cleaving enzyme 1 (BACE1) thereby resulting in increased secretion of amyloid β peptides (Aβ1-40 and Aβ1-42). Furthermore, increased expression of APP and BACE1 as well as increased production of Aβ peptides was detected in the cerebral microvasculature and brain tissue of eNOS-deficient mice. Since Aβ peptides are considered major cytotoxic molecules responsible for the pathogenesis of Alzheimers 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.


Journal of Neurochemistry | 2013

Endothelial nitric oxide deficiency promotes Alzheimer's disease pathology.

Susan A. Austin; Anantha Vijay R. Santhanam; David J. Hinton; Doo Sup Choi; Zvonimir S. Katusic

Aging and the presence of cerebrovascular disease are associated with increased incidence of Alzheimers 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, β‐site APP cleaving enzyme (BACE) 1, and amyloid beta (Aβ) levels were significantly higher in the brains of LMA eNOS−/− mice as compared with LMA wild‐type controls. APP and Aβ1‐40 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 Aβ related pathology and cognitive decline.


Circulation | 2016

Neurovascular Protective Function of Endothelial Nitric Oxide – Recent Advances –

Zvonimir S. Katusic; Susan A. Austin

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 Alzheimers 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).


Circulation Research | 2016

Loss of Endothelial Nitric Oxide Synthase Promotes p25 Generation and Tau Phosphorylation in a Murine Model of Alzheimer’s Disease

Susan A. Austin; Zvonimir S. Katusic

RATIONALE Alzheimers disease has an unknown pathogenesis; however, cardiovascular risk factors are associated with a higher incidence of Alzheimers 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 Alzheimers 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 Alzheimers disease.


Journal of Alzheimer's Disease | 2012

Supplementation of Nitric Oxide Attenuates AβPP and BACE1 Protein in Cerebral Microcirculation of eNOS-Deficient Mice

Susan A. Austin; Livius V. d'Uscio; Zvonimir S. Katusic

Recently, we demonstrated in endothelial nitric oxide synthase deficient (eNOS-/-) mice that loss of endothelial NO led to increased protein levels of amyloid-β protein precursor (AβPP), β-site AβPP cleaving enzyme 1 (BACE1), and amyloid-β (Aβ) peptide. Therefore, our aim was to determine if NO supplementation in vivo would attenuate AβPP and BACE1 protein levels. cGMP levels were significantly increased while AβPP 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 AβPP.


PLOS ONE | 2015

Regional Heterogeneity of Cerebral Microvessels and Brain Susceptibility to Oxidative Stress

Susan A. Austin; Anantha Vijay R. Santhanam; Livius V. d’Uscio; Zvonimir S. Katusic

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.


PLOS ONE | 2015

Characterization of Novel Src Family Kinase Inhibitors to Attenuate Microgliosis.

Gunjan D. Manocha; Kendra L. Puig; Susan A. Austin; Kathleen Seyb; Marcie A. Glicksman; Colin K. Combs

Microgliosis is a major hallmark of Alzheimer’s disease (AD) brain pathology. Aβ 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 Aβ-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 Aβ-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 Aβ 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.


PLOS ONE | 2015

Intracellular superoxide anions are increased in the microvessels isolated from the cortex and hippocampus.

Susan A. Austin; Anantha Vijay R. Santhanam; Livius V. d’Uscio; Zvonimir S. Katusic


PLOS ONE | 2015

Protein levels of COX-1 and PGI-2S did not differ between brain region microvessels.

Susan A. Austin; Anantha Vijay R. Santhanam; Livius V. d’Uscio; Zvonimir S. Katusic

Collaboration


Dive into the Susan A. Austin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Colin K. Combs

University of North Dakota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gunjan D. Manocha

University of North Dakota

View shared research outputs
Top Co-Authors

Avatar

Kathleen Seyb

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Kendra L. Puig

University of North Dakota

View shared research outputs
Researchain Logo
Decentralizing Knowledge