Network


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

Hotspot


Dive into the research topics where John E. Donahue is active.

Publication


Featured researches published by John E. Donahue.


Acta Neuropathologica | 2006

RAGE, LRP-1, and amyloid-beta protein in Alzheimer’s disease

John E. Donahue; Stephanie Flaherty; Conrad E. Johanson; John Duncan; Gerald D. Silverberg; Miles C. Miller; Rosemarie Tavares; Wentian Yang; Qian Wu; Edmond Sabo; Virginia Hovanesian; Edward G. Stopa

The receptor for advanced glycation end products (RAGE) is thought to be a primary transporter of β-amyloid across the blood–brain barrier (BBB) into the brain from the systemic circulation, while the low-density lipoprotein receptor-related protein (LRP)-1 mediates transport of β-amyloid out of the brain. To determine whether there are Alzheimer’s disease (AD)-related changes in these BBB-associated β-amyloid receptors, we studied RAGE, LRP-1, and β-amyloid in human elderly control and AD hippocampi. In control hippocampi, there was robust RAGE immunoreactivity in neurons, whereas microvascular staining was barely detectable. LRP-1 staining, in contrast, was clearly evident within microvessels but only weakly stained neurons. In AD cases, neuronal RAGE immunoreactivity was significantly decreased. An unexpected finding was the strongly positive microvascular RAGE immunoreactivity. No evidence for colocalization of RAGE and β-amyloid was seen within either microvessels or senile plaques. A reversed pattern was evident for LRP-1 in AD. There was very strong staining for LRP-1 in neurons, with minimal microvascular staining. Unlike RAGE, colocalization of LRP-1 and β-amyloid was clearly present within senile plaques but not microvessels. Western blot analysis revealed a much higher concentration of RAGE protein in AD hippocampi as compared with controls. Concentration of LRP-1 was increased in AD hippocampi, likely secondary to its colocalization with senile plaques. These data confirm that AD is associated with changes in the relative distribution of RAGE and LRP-1 receptors in human hippocampus. They also suggest that the proportion of amyloid within the brains of AD patients that is derived from the systemic circulation may be significant.


Journal of Neuropathology and Experimental Neurology | 2010

Amyloid efflux transporter expression at the blood-brain barrier declines in normal aging.

Gerald D. Silverberg; Arthur Messier; Miles C. Miller; Jason T. Machan; Samir Majmudar; Edward G. Stopa; John E. Donahue; Conrad E. Johanson

Reduced clearance of amyloid &bgr; peptides (A&bgr;) across the blood-brain barrier contributes to amyloid accumulation in Alzheimer disease. Amyloid &bgr; efflux transport is via the endothelial low-density lipoprotein receptor-related protein 1 (LRP-1) and P-glycoprotein (P-gp), whereas A&bgr; influx transport is via the receptor for advanced glycation end products. Because age is the major risk factor for developing Alzheimer disease, we measured LRP-1 and P-gp expression and associated transporter expression with A&bgr; accumulation in aging rats. Quantitative LRP-1 and P-gp microvessel expression was measured by immunohistochemistry (IHC); LRP-1 and P-gp expression were assessed in microvessel isolates by Western blotting. There was an age-dependent loss of capillary LRP-1 across all ages (3-36 months) by IHC (linear trend p = 0.0004) and between 3 and 20 months by Western blotting (linear trend p < 0.0001). There was a late (30-36 months) P-gp expression loss by IHC (p < 0.05) and Western blotting (p = 0.0112). Loss ofLRP-1 correlated with A&bgr;42 accumulation (p = 0.0121) and verynearly with A&bgr;40 (p = 0.0599) across all ages. Expression of LRP-1correlated negatively with the expression of receptor for advanced glycation end products (p < 0.0004). These data indicate that alterations in LRP-1 and P-gp expression seem to contribute progressively to A&bgr; accumulation in aging.


Brain Research | 2008

Hippocampal RAGE Immunoreactivity in Early and Advanced Alzheimer’s Disease

Miles C. Miller; Rosemarie Tavares; Conrad E. Johanson; Virginia Hovanesian; John E. Donahue; Liliana Gonzalez; Gerald D. Silverberg; Edward G. Stopa

Microvascular accumulation and neuronal overproduction of amyloid-beta peptide (Abeta) are pathologic features of Alzheimers disease (AD). In this study, we examined the receptor for advanced glycation endproducts (RAGE), a multi-ligand receptor found in both neurons and cerebral microvascular endothelia that binds Abeta. RAGE expression was assessed in aged controls (n = 6), patients with early AD-like pathology (n = 6), and severe, Braak V-VI AD (n = 6). Human hippocampi were stained with a specific polyclonal antibody directed against RAGE (Research Diagnostics, Flanders, NJ). Immunoreactivity was localized in both neurons and cerebral endothelial cells. Quantitative image-analyses were performed on grayscale images to assess the total surface area of endothelial RAGE immunoreaction product in cross sections of cerebral microvessels (5-20 microm). Confocal images were acquired for confirmation of RAGE immunoreactivity in both microvessels and neurons by coupling RAGE with CD-31 and neurofilament, respectively. A significant increase in endothelial RAGE immunoreactivity was found in severe Braak V-VI AD patients when compared to aged controls (p < 0.001), and when compared to patients with early AD pathology (p = 0.0125). In addition, a significant increase in endothelial RAGE immunoreactivity was witnessed when comparing aged controls having no reported AD pathology with patients having early AD-like pathology (p = 0.038). Our data suggest that microvascular RAGE levels increase in conjunction with the onset of AD, and continue to increase linearly as a function of AD pathologic severity (p < 0.0001).


Brain Research | 2000

Cells containing immunoreactive estrogen receptor-α in the human basal forebrain

John E. Donahue; Edward G. Stopa; Regina L Chorsky; Joan C. King; Hyman M. Schipper; Stuart A. Tobet; Jeffrey D. Blaustein; Seymour Reichlin

The distribution of estrogen receptor protein-α (ER-α)-containing cells in the human hypothalamus and adjacent regions was studied using a monoclonal antibody (H222) raised against ER-α derived from MCF-7 human breast cancer cells. Reaction product was found in restricted populations of neurons and astrocyte-like cells. Neurons immunoreactive for ER-α were diffusely distributed within the basal forebrain and preoptic area, infundibular region, central hypothalamus, basal ganglia and amygdala. Immunoreactive astrocyte-like cells were noted within specific brain regions, including the lamina terminalis and subependymal peri-third-ventricular region. These data are consistent with the location of estrogen receptors in the basal forebrain of other species and the known effects of estrogens on the cellular functions of both neurons and supporting elements within the human hypothalamus and basal forebrain.


Journal of Neuropathology and Experimental Neurology | 2010

Amyloid deposition and influx transporter expression at the blood-brain barrier increase in normal aging.

Gerald D. Silverberg; Miles C. Miller; Arthur Messier; Samir Majmudar; Jason T. Machan; John E. Donahue; Edward G. Stopa; Conrad E. Johanson

Aging is the most important single risk factor for developing Alzheimer disease. We measured amyloid-&bgr; peptide (A&bgr;) levels in rat cerebral cortex and hippocampus during normal aging of Brown-Norway/Fischer rats. Amyloid-&bgr; accumulation was associated with expression of the A&bgr; influx transporter, the receptor for advanced glycation end-products (RAGEs) at the blood-brain barrier. Rats at selected ages from 3 to 36 months were analyzed by 1) immunohistochemistry for amyloid deposition and quantitative microvessel surfacearea RAGE expression, 2) ELISA for cortical A&bgr;40 and A&bgr;42 concentrations, and 3) Western blotting of microvessel proteins for RAGE expression. Immunohistochemistry showed increasing accumulation of brain A&bgr; with aging. By ELISA analysis, both A&bgr;40 and A&bgr;42 concentrations in cortical homogenates rose sharply from 9 to 12 months. The A&bgr;42 continued to rise up to age 30 months, whereas A&bgr;40 stabilized after 12 months. The expression of RAGE initially decreased between 3 and 12 months but then increased between 12 and 34 months by immunohistochemistry. On immunoblotting, RAGE decreased up to 9 months and then progressively increased up to 36 months. These data indicate an association between amyloid and microvessel RAGE during aging. An increase in capillary RAGE expression seems to play a role in the later A&bgr; accumulation but not in the initial increase.


Journal of Neuropathology and Experimental Neurology | 2008

Apolipoprotein E, Amyloid-β, and Blood-Brain Barrier Permeability in Alzheimer Disease

John E. Donahue; Conrad E. Johanson

There is increasing evidence for blood-brain barrier (BBB) compromise in Alzheimer disease (AD). The presence of the ϵ4 allele of the apolipoprotein E (apoE) gene is a risk factor for sporadic AD. Apolipoprotein E is essential both for maintenance of BBB integrity and for the deposition of fibrillar amyloid-&bgr; (A&bgr;) that leads to the development of A&bgr; plaques in AD and to cerebral amyloid angiopathy. This review investigates the relationships between apoE, A&bgr;, and the BBB in AD. Alterations in the expression and distribution of the BBB A&bgr; transporters receptor for advanced glycation end-products and low-density lipoprotein receptor-related protein 1 in AD and the potential roles of apoE4 expression in adversely influencing A&bgr; burden and BBB permeability are also examined. Because both apoE and A&bgr; are ligands for low-density lipoprotein receptor-related protein 1, all 3 molecules are present in AD plaques, and most AD plaques are located close to the cerebral microvasculature. The interactions of these molecules at the BBB likely influence metabolism and clearance of A&bgr; and contribute to AD pathogenesis. Therapeutic alternatives targeting apoE/A&bgr; and sealing a compromised BBB are under development for the treatment of AD.


Journal of Alzheimer's Disease | 2010

Brain ventricular volume and cerebrospinal fluid biomarkers of Alzheimer’s disease

Brian R. Ott; Ronald A. Cohen; Assawin Gongvatana; Ozioma C. Okonkwo; Conrad E. Johanson; Edward G. Stopa; John E. Donahue; Gerald D. Silverberg

The frequent co-occurrence of Alzheimers disease (AD) pathology in patients with normal pressure hydrocephalus suggests a possible link between ventricular dilation and AD. If enlarging ventricles serve as a marker of faulty cerebrospinal fluid (CSF) clearance mechanisms, then a relationship may be demonstrable between increasing ventricular volume and decreasing levels of amyloid-beta peptide (Abeta) in CSF in preclinical and early AD. CSF biomarker data (Abeta, tau, and phosphorylated tau) as well as direct measurements of whole brain and ventricular volumes were obtained from the Alzheimers Disease Neuroimaging Initiative dataset. The ratio of ventricular volume to whole brain volume was derived as a secondary independent measure. Baseline data were used for the group analyses of 288 subjects classified as being either normal (n=87), having the syndrome of mild cognitive impairment (n=136), or mild AD (n=65). Linear regression models were derived for each biomarker as the dependent variable, using the MRI volume measures and age as independent variables. For controls, ventricular volume was negatively associated with CSF Abeta in APOE epsilon4 positive subjects. A different pattern was seen in AD subjects, in whom ventricular volume was negatively associated with tau, but not Abeta in epsilon4 positive subjects. Increased ventricular volume may be associated with decreased levels of CSF Abeta in preclinical AD. The basis for the apparent effect of APOE epsilon4 genotype on the relationship of ventricular volume to Abeta and tau levels is unknown, but could involve altered CSF-blood-brain barrier function during the course of disease.


Fluids and Barriers of the CNS | 2011

Ecrg4 expression and its product augurin in the choroid plexus: impact on fetal brain development, cerebrospinal fluid homeostasis and neuroprogenitor cell response to CNS injury

Ana Maria Gonzalez; Sonia Podvin; Shuh-Yow Lin; Miles C. Miller; Hannah Botfield; Wendy Leadbeater; Andrew Roberton; Xitong Dang; Stuart Knowling; Elena Cardenas-Galindo; John E. Donahue; Edward G. Stopa; Conrad E. Johanson; Raul Coimbra; Brian P. Eliceiri; Andrew Baird

BackgroundThe content and composition of cerebrospinal fluid (CSF) is determined in large part by the choroid plexus (CP) and specifically, a specialized epithelial cell (CPe) layer that responds to, synthesizes, and transports peptide hormones into and out of CSF. Together with ventricular ependymal cells, these CPe relay homeostatic signals throughout the central nervous system (CNS) and regulate CSF hydrodynamics. One new candidate signal is augurin, a newly recognized 14 kDa protein that is encoded by esophageal cancer related gene-4 (Ecrg4), a putative tumor suppressor gene whose presence and function in normal tissues remains unexplored and enigmatic. The aim of this study was to explore whether Ecrg4 and its product augurin, can be implicated in CNS development and the response to CNS injury.MethodsEcrg4 gene expression in CNS and peripheral tissues was studied by in situ hybridization and quantitative RT-PCR. Augurin, the protein encoded by Ecrg4, was detected by immunoblotting, immunohistochemistry and ELISA. The biological consequence of augurin over-expression was studied in a cortical stab model of rat CNS injury by intra-cerebro-ventricular injection of an adenovirus vector containing the Ecrg4 cDNA. The biological consequences of reduced augurin expression were evaluated by characterizing the CNS phenotype caused by Ecrg4 gene knockdown in developing zebrafish embryos.ResultsGene expression and immunohistochemical analyses revealed that, the CP is a major source of Ecrg4 in the CNS and that Ecrg4 mRNA is predominantly localized to choroid plexus epithelial (CPe), ventricular and central canal cells of the spinal cord. After a stab injury into the brain however, both augurin staining and Ecrg4 gene expression decreased precipitously. If the loss of augurin was circumvented by over-expressing Ecrg4 in vivo, BrdU incorporation by cells in the subependymal zone decreased. Inversely, gene knockdown of Ecrg4 in developing zebrafish embryos caused increased proliferation of GFAP-positive cells and induced a dose-dependent hydrocephalus-like phenotype that could be rescued by co-injection of antisense morpholinos with Ecrg4 mRNA.ConclusionAn unusually elevated expression of the Ecrg4 gene in the CP implies that its product, augurin, plays a role in CP-CSF-CNS function. The results are all consistent with a model whereby an injury-induced decrease in augurin dysinhibits target cells at the ependymal-subependymal interface. We speculate that the ability of CP and ependymal epithelium to alter the progenitor cell response to CNS injury may be mediated, in part by Ecrg4. If so, the canonic control of its promoter by DNA methylation may implicate epigenetic mechanisms in neuroprogenitor fate and function in the CNS.


American Journal of Roentgenology | 2010

Preoperative MRI evaluation of pituitary macroadenoma: imaging features predictive of successful transsphenoidal surgery.

Jerrold L. Boxerman; Jeffrey M. Rogg; John E. Donahue; Jason T. Machan; Marc A. Goldman; Curt E. Doberstein

OBJECTIVE The purpose of this study was to determine whether the preoperative MRI findings of enhanced diffusivity, macrocyst content, and internal hemorrhage in pituitary macroadenomas are predictive of successful transsphenoidal hypophysectomy. MATERIALS AND METHODS We retrospectively reviewed the preoperative and postoperative sella protocol MR images of 28 patients who underwent transsphenoidal hypophysectomy for chiasm-compressing macroadenoma. Chiasmatic decompression defined surgical success. Two neuroradiologists differentiated nonsolid (macrocystic and macrohemorrhagic) from solid tumors, computed apparent diffusion coefficient (ADC) and T2-weighted signal intensity normalized to pons in solid tumors, and measured change in tumor height. A neuropathologist graded reticulin content in tumor specimens. Categorical and dichotomous variables were examined with the chi-square or Fishers exact test; continuous-scale data were analyzed with the Students t test, analysis of variance, or linear regression. RESULTS Transsphenoidal hypophysectomy succeeded in the management of 10 of 11 nonsolid tumors and nine of 17 solid tumors (p = 0.049). The ratios of tumor to brainstem ADC in the nine successfully resected solid tumors were higher than in the eight cases of failed treatment (p = 0.008) with no significant difference in ratio of tumor to brainstem T2-weighted signal intensity (p = 0.76). All six solid tumors with enhanced diffusivity (ratio of tumor to brainstem ADC > 1.1) were successfully managed with transsphenoidal hypophysectomy, compared with three of 11 with an ADC ratio less than 1.1 (p = 0.009). There was a significant main effect of ADC ratio groupings on change in tumor height (p = 0.02), and a linear relation was found between ADC ratio and change in tumor height (p = 0.04). Taken together, tumors with nonsolid features or an ADC ratio greater than 1.1 were highly resectable (p < 0.001; sensitivity, 0.84; specificity, 0.89). ADC ratios in reticulin-poor solid tumors were higher than those in reticulin-rich tumors (p = 0.024). CONCLUSION Macrocystic and macrohemorrhagic adenomas and solid tumors with enhanced diffusivity are more likely to be successfully managed with transsphenoidal hypophysectomy. Transsphenoidal hypophysectomy of solid, enhancing tumors with restricted diffusion is more likely to fail, possibly because of the greater reticulin content of the tumor; initial transcranial surgery may be appropriate in these cases.


Acta Neuropathologica | 1998

Intracerebral hemorrhage in two patients with Down's syndrome and cerebral amyloid angiopathy.

John E. Donahue; Jasvir S. Khurana; Lester S. Adelman

Abstract Cerebral amyloid angiopathy (CAA) is an important cause of spontaneous intracerebral hemorrhages in the elderly and is often seen in the brains of patients with Alzheimer’s disease, Down’s syndrome (DS), and hereditary cerebral hemorrhage with amyloidosis of the Dutch type. We report two patients with DS and extensive CAA who died of intracerebral hemorrhage; only two other such case reports exist in the literature. We believe the incidence of such cases is higher than is reported and that the likelihood of hemorrhage in the setting of CAA is independent of the patient’s underlying disease.

Collaboration


Dive into the John E. Donahue's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sonia Podvin

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge