Network


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

Hotspot


Dive into the research topics where Adviye Ergul is active.

Publication


Featured researches published by Adviye Ergul.


Cardiovascular Diabetology | 2005

Oxidative stress and the use of antioxidants in diabetes: Linking basic science to clinical practice

Jeanette Schultz Johansen; Alex K. Harris; David J. Rychly; Adviye Ergul

Cardiovascular complications, characterized by endothelial dysfunction and accelerated atherosclerosis, are the leading cause of morbidity and mortality associated with diabetes. There is growing evidence that excess generation of highly reactive free radicals, largely due to hyperglycemia, causes oxidative stress, which further exacerbates the development and progression of diabetes and its complications. Overproduction and/or insufficient removal of these free radicals result in vascular dysfunction, damage to cellular proteins, membrane lipids and nucleic acids. Despite overwhelming evidence on the damaging consequences of oxidative stress and its role in experimental diabetes, large scale clinical trials with classic antioxidants failed to demonstrate any benefit for diabetic patients. As our understanding of the mechanisms of free radical generation evolves, it is becoming clear that rather than merely scavenging reactive radicals, a more comprehensive approach aimed at preventing the generation of these reactive species as well as scavenging may prove more beneficial. Therefore, new strategies with classic as well as new antioxidants should be implemented in the treatment of diabetes.


Stroke | 2010

Minocycline to Improve Neurologic Outcome in Stroke (MINOS): A dose-finding study

Susan C. Fagan; Jennifer L. Waller; Fenwick T. Nichols; David J. Edwards; L. Creed Pettigrew; Wayne M. Clark; Christiana E. Hall; Jeffrey A. Switzer; Adviye Ergul; David C. Hess

Background and Purpose— Minocycline is a promising anti-inflammatory and protease inhibitor that is effective in multiple preclinical stroke models. We conducted an early phase trial of intravenous minocycline in acute ischemic stroke. Methods— Following an open-label, dose-escalation design, minocycline was administered intravenously within 6 hours of stroke symptom onset in preset dose tiers of 3, 4.5, 6, or 10 mg/kg daily over 72 hours. Minocycline concentrations for pharmacokinetic analysis were measured in a subset of patients. Subjects were followed for 90 days. Results— Sixty patients were enrolled, 41 at the highest dose tier of 10 mg/kg. Overall age (65±13.7 years), race (83% white), and sex (47% female) were consistent across the doses. The mean baseline National Institutes of Health Stroke Scale score was 8.5±5.8 and 60% received tissue plasminogen activator. Minocycline infusion was well tolerated with only 1 dose limiting toxicity at the 10-mg/kg dose. No severe hemorrhages occurred in tissue plasminogen activator-treated patients. Pharmacokinetic analysis (n=22) revealed a half-life of approximately 24 hours and linearity of parameters over doses. Conclusions— Minocycline is safe and well tolerated up to doses of 10 mg/kg intravenously alone and in combination with tissue plasminogen activator. The half-life of minocycline is approximately 24 hours, allowing every 24-hour dosing. Minocycline may be an ideal agent to use with tissue plasminogen activator.


BMC Neuroscience | 2006

Delayed minocycline inhibits ischemia-activated matrix metalloproteinases 2 and 9 after experimental stroke

Livia S. Machado; Anna Kozak; Adviye Ergul; David C. Hess; Cesario V. Borlongan; Susan C. Fagan

BackgroundMatrix metalloproteinases 2 and 9 (MMP-2 and MMP-9) are increased in the brain after experimental ischemic stroke in rats. These two proteases are involved with the degradation of the basal lamina and loss of stability of the blood brain barrier that occurs after ischemia and that is associated with thrombolytic therapy in ischemic stroke. Minocycline is a lipophilic tetracycline and is neuroprotective in several models of brain injury. Minocycline inhibits inflammation, apoptosis and extracellular matrix degradation. In this study we investigated whether delayed minocycline inhibits brain MMPs activated by ischemia in a model of temporary occlusion in Wistar rats.ResultsBoth MMP-2 and MMP-9 were elevated in the ischemic tissue as compared to the contra-lateral hemisphere after 3 hours occlusion and 21 hours survival (p < 0.0001 for MMP-9). Intraperitoneal minocycline at 45 mg/kg concentration twice a day (first dose immediately after the onset of reperfusion) significantly reduced gelatinolytic activity of ischemia-elevated MMP-2 and MMP-9 (p < 0.0003). Treatment also reduced protein concentration of both enzymes (p < 0.038 for MMP-9 and p < 0.018 for MMP-2). In vitro incubation of minocycline in concentrations as low as 0.1 μg/ml with recombinant MMP-2 and MMP-9 impaired enzymatic activity and MMP-9 was more sensitive at lower minocycline concentrations (p < 0.05).ConclusionMinocycline inhibits enzymatic activity of gelatin proteases activated by ischemia after experimental stroke and is likely to be selective for MMP-9 at low doses. Minocycline is a potential new therapeutic agent to acute treatment of ischemic stroke.


Stroke | 2012

Angiogenesis A Harmonized Target for Recovery After Stroke

Adviye Ergul; Ahmed Alhusban; Susan C. Fagan

It is now appreciated that emerging therapeutic strategies for recovery must include the cerebral vasculature and that induction of angiogenesis will stimulate endogenous recovery mechanisms, including neurogenesis, synaptogenesis, and neuronal and synaptic plasticity. These events are all involved in the long-term repair and restoration process of the brain after an ischemic event. Several recent excellent reviews provided detailed information on the mechanisms and molecular targets for angiogenesis after stroke.1,2 The purpose of this review is to evaluate the evidence that angiogenesis is a target for recovery after an ischemic stroke. ### Angiogenic Response to Ischemic Brain Injury: A Multipurpose Pathway Early reports of increased angiogenesis in the ischemic border zone of human brain autopsy sections,3 which was decreased in patients of advanced age,4 led to interest in the time course and impact of this phenomenon on functional recovery. It is clear that angiogenesis genes are upregulated within minutes of the onset of cerebral ischemia in rodents5 and angiogenic proteins remain increased in the area of ischemia for days to weeks.6 It is unclear, however, whether the angiogenic response leads to the development of functional new blood vessels that improve brain function after stroke. Clinical and experimental studies in other vascular beds have emphasized the potential for adverse consequences related to neovascularization.7,8 In the diabetic retina, for example, pathological angiogenesis results in hemorrhage, edema, and, ultimately, blindness.9 In the brain, pathological angiogenesis is implicated in the development of hereditary hemorrhagic telangiectasia.10 The correlation between angiogenesis and improved functional outcome after ischemic stroke remains and is seen in both animal models and in human patients with stroke.5,11–13 It is likely that the “proangiogenic state,” induced in response to an ischemic insult, has multiple purposes in the hours to weeks after the injury (Figure). First, the …


Pharmacological Research | 2011

Endothelin-1 and diabetic complications: Focus on the vasculature

Adviye Ergul

Diabetes is not only an endocrine but also a vascular disease. Cardiovascular complications are the leading cause of morbidity and mortality associated with diabetes. Diabetes affects both large and small vessels and hence diabetic complications are broadly classified as microvascular (retinopathy, nephropathy and neuropathy) and macrovascular (heart disease, stroke and peripheral arterial disease) complications. Endothelial dysfunction, defined as an imbalance of endothelium-derived vasoconstrictor and vasodilator substances, is a common denominator in the pathogenesis and progression of both macro and microvascular complications. While the pathophysiology of diabetic complications is complex, endothelin-1 (ET-1), a potent vasoconstrictor with proliferative, profibrotic, and proinflammatory properties, may contribute to many facets of diabetic vascular disease. This review will focus on the effects of ET-1 on function and structure of microvessels (retina, skin and mesenteric arteries) and macrovessels (coronary and cerebral arteries) and also discuss the relative role(s) of endothelin A (ET(A)) and ET(B) receptors in mediating ET-1 actions.


BMC Neurology | 2007

Increased hemorrhagic transformation and altered infarct size and localization after experimental stroke in a rat model type 2 diabetes

Adviye Ergul; Mostafa M. Elgebaly; Mary Louise Middlemore; Weiguo Li; Hazem Elewa; Jeffrey A. Switzer; Christiana E. Hall; Anna Kozak; Susan C. Fagan

BackgroundInterruption of flow through of cerebral blood vessels results in acute ischemic stroke. Subsequent breakdown of the blood brain barrier increases cerebral injury by the development of vasogenic edema and secondary hemorrhage known as hemorrhagic transformation (HT). Diabetes is a risk factor for stroke as well as poor outcome of stroke. The current study tested the hypothesis that diabetes-induced changes in the cerebral vasculature increase the risk of HT and augment ischemic injury.MethodsDiabetic Goto-Kakizaki (GK) or control rats underwent 3 hours of middle cerebral artery occlusion and 21 h reperfusion followed by evaluation of infarct size, hemorrhage and neurological outcome.ResultsInfarct size was significantly smaller in GK rats (10 ± 2 vs 30 ± 4%, p < 0.001). There was significantly more frequent hematoma formation in the ischemic hemisphere in GK rats as opposed to controls. Cerebrovascular tortuosity index was increased in the GK model (1.13 ± 0.01 vs 1.34 ± 0.06, P < 0.001) indicative of changes in vessel architecture.ConclusionThese findings provide evidence that there is cerebrovascular remodeling in diabetes. While diabetes-induced remodeling appears to prevent infarct expansion, these changes in blood vessels increase the risk for HT possibly exacerbating neurovascular damage due to cerebral ischemia/reperfusion in diabetes.


Stroke | 2011

Matrix Metalloproteinase-9 in an Exploratory Trial of Intravenous Minocycline for Acute Ischemic Stroke

Jeffrey A. Switzer; David C. Hess; Adviye Ergul; Jennifer L. Waller; Livia S. Machado; Vera Portik-Dobos; L. Creed Pettigrew; Wayne M. Clark; Susan C. Fagan

Background and Purpose— Plasma matrix metalloproteinase-9 levels predict posttissue plasminogen activator (tPA) hemorrhage. Methods— The authors investigated the effect of minocycline on plasma matrix metalloproteinase-9 in acute ischemic stroke in the Minocycline to Improve Neurological Outcome in Stroke (MINOS) trial and a comparison group. Results— Matrix metalloproteinase-9 level decreased at 72 hours compared with baseline in MINOS (tPA, P=0.0022; non-tPA, P=0.0066) and was lower than in the non-MINOS comparison group at 24 hours (tPA, P<0.0001; non-tPA, P=0.0019). Conclusions— Lower plasma matrix metalloproteinase-9 was seen among tPA-treated subjects in the MINOS trial. Combining minocycline with tPA may prevent the adverse consequences of thrombolytic therapy through suppression of matrix metalloproteinase-9 activity.


Stroke | 2009

Minocycline and Tissue-Type Plasminogen Activator for Stroke Assessment of Interaction Potential

Livia S. Machado; Irina Y. Sazonova; Anna Kozak; Daniel C. Wiley; Azza B. El-Remessy; Adviye Ergul; David C. Hess; Jennifer L. Waller; Susan C. Fagan

Background and Purpose— New treatment strategies for acute ischemic stroke must be evaluated in the context of effective reperfusion. Minocycline is a neuroprotective agent that inhibits proteolytic enzymes and therefore could potentially both inactivate the clot lysis effect and decrease the damaging effects of tissue-type plasminogen activator (t-PA). This study aimed to determine the effect of minocycline on t-PA clot lysis and t-PA–induced hemorrhage formation after ischemia. Methods— Fibrinolytic and amidolytic activities of t-PA were investigated in vitro over a range of clinically relevant minocycline concentrations. A suture occlusion model of 3-hour temporary cerebral ischemia in rats treated with t-PA and 2 different minocycline regimens was used. Blood–brain barrier basal lamina components, matrix metalloproteinases (MMPs), hemorrhage formation, infarct size, edema, and behavior outcome were assessed. Results— Minocycline did not affect t-PA fibrinolysis. However, minocycline treatment at 3 mg/kg IV decreased total protein expression of both MMP-2 (P=0.0034) and MMP-9 (P=0.001 for 92 kDa and P=0.0084 for 87 kDa). It also decreased the incidence of hemorrhage (P=0.019), improved neurologic outcome (P=0.0001 for Bederson score and P=0.0391 for paw grasp test), and appeared to decrease mortality. MMP inhibition was associated with decreased degradation in collagen IV and laminin-α1 (P=0.0001). Conclusions— Combination treatment with minocycline is beneficial in t-PA–treated animals and does not compromise clot lysis. These results also suggest that neurovascular protection by minocycline after stroke may involve direct protection of the blood–brain barrier during thrombolysis with t-PA.


Vascular Pharmacology | 2009

Hyperglycemia, diabetes and stroke: Focus on the cerebrovasculature

Adviye Ergul; Weiguo Li; Mostafa M. Elgebaly; Askiel Bruno; Susan C. Fagan

Acute ischemic stroke (AIS) results from the occlusion of an artery and causes vascular and neuronal damage, both of which affect the extent of ischemic injury and stroke outcome. Despite extensive efforts, there is only one effective treatment for AIS. Given that up to 40% of the AIS patients present with admission hyperglycemia either as a result of diabetes or acute stress response, targets for neuronal and vascular protection under hyperglycemic conditions need to be better defined. Here, we review the impact of diabetes and acute hyperglycemia on experimental stroke with an emphasis on cerebrovasculature structure and function. The relevance to clinical evidence is also discussed.


BMC Neuroscience | 2005

Effect of neutrophil depletion on gelatinase expression, edema formation and hemorrhagic transformation after focal ischemic stroke

Alex K. Harris; Adviye Ergul; Anna Kozak; Livia S. Machado; Maribeth H. Johnson; Susan C. Fagan

BackgroundWhile gelatinase (MMP-2 and -9) activity is increased after focal ischemia/reperfusion injury in the brain, the relative contribution of neutrophils to the MMP activity and to the development of hemorrhagic transformation remains unknown.ResultsAnti-PMN treatment caused successful depletion of neutrophils in treated animals. There was no difference in either infarct volume or hemorrhage between control and PMN depleted animals. While there were significant increases in gelatinase (MMP-2 and MMP-9) expression and activity and edema formation associated with ischemia, neutrophil depletion failed to cause any change.ConclusionThe main finding of this study is that, in the absence of circulating neutrophils, MMP-2 and MMP-9 expression and activity are still up-regulated following focal cerebral ischemia. Additionally, neutrophil depletion had no influence on indicators of ischemic brain damage including edema, hemorrhage, and infarct size. These findings indicate that, at least acutely, neutrophils are not a significant contributor of gelatinase activity associated with acute neurovascular damage after stroke.

Collaboration


Dive into the Adviye Ergul's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Weiguo Li

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maha Coucha

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tauheed Ishrat

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge