Network


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

Hotspot


Dive into the research topics where Jeffrey N. Keller is active.

Publication


Featured researches published by Jeffrey N. Keller.


Journal of Neurochemistry | 2009

Morphine and HIV-Tat increase microglial-free radical production and oxidative stress: possible role in cytokine regulation

Jadwiga Turchan-Cholewo; Filomena O. Dimayuga; Sunita Gupta; Jeffrey N. Keller; Pamela E. Knapp; Kurt F. Hauser; Annadora J. Bruce-Keller

Opiate abuse alters the progression of human immunodeficiency virus and may increase the risk of neuroAIDS. As neuroAIDS is associated with altered microglial reactivity, the combined effects of human immunodeficiency virus‐Tat and morphine were determined in cultured microglia. Specifically, experiments determined the effects of Tat and morphine on microglial‐free radical production and oxidative stress, and on cytokine release. Data show that combined Tat and morphine cause early and synergistic increases in reactive oxygen species, with concomitant increases in protein oxidation. Furthermore, combined Tat and morphine, but not Tat or morphine alone, cause reversible decreases in proteasome activity. The effects of morphine on free radical production and oxidative stress are prevented by pre‐treatment with naloxone, illustrating the important role of opioid receptor activation in these phenomena. While Tat is well known to induce cytokine release from cultured microglia, morphine decreases Tat‐induced release of the cytokines tumor necrosis factor‐α and interleukin‐6, as well as the chemokine monocyte chemoattractant protein‐1 (MCP‐1). Finally, experiments using the reversible proteasome inhibitor MG115 show that temporary, non‐cytotoxic decreases in proteasome activity increase protein oxidation and decrease tumor necrosis factor‐α, interleukin‐6, and MCP‐1 release from microglia. Taken together, these data suggest that oxidative stress and proteasome inhibition may be involved in the immunomodulatory properties of opioid receptor activation in microglia.


Neuropharmacology | 2014

Differences in amyloid-β clearance across mouse and human blood–brain barrier models: Kinetic analysis and mechanistic modeling

Hisham Qosa; Bilal S. Abuasal; Ignacio A. Romero; Babette B. Weksler; Pierre-Oliver Couraud; Jeffrey N. Keller; Amal Kaddoumi

Alzheimers disease (AD) has a characteristic hallmark of amyloid-β (Aβ) accumulation in the brain. This accumulation of Aβ has been related to its faulty cerebral clearance. Indeed, preclinical studies that used mice to investigate Aβ clearance showed that efflux across blood-brain barrier (BBB) and brain degradation mediate efficient Aβ clearance. However, the contribution of each process to Aβ clearance remains unclear. Moreover, it is still uncertain how species differences between mouse and human could affect Aβ clearance. Here, a modified form of the brain efflux index method was used to estimate the contribution of BBB and brain degradation to Aβ clearance from the brain of wild type mice. We estimated that 62% of intracerebrally injected (125)I-Aβ40 is cleared across BBB while 38% is cleared by brain degradation. Furthermore, inxa0vitro and in silico studies were performed to compare Aβ clearance between mouse and human BBB models. Kinetic studies for Aβ40 disposition in bEnd3 and hCMEC/D3 cells, representative inxa0vitro mouse and human BBB models, respectively, demonstrated 30-fold higher rate of (125)I-Aβ40 uptake and 15-fold higher rate of degradation by bEnd3 compared to hCMEC/D3 cells. Expression studies showed both cells to express different levels of P-glycoprotein and RAGE, while LRP1 levels were comparable. Finally, we established a mechanistic model, which could successfully predict cellular levels of (125)I-Aβ40 and the rate of each process. Established mechanistic model suggested significantly higher rates of Aβ uptake and degradation in bEnd3 cells as rationale for the observed differences in (125)I-Aβ40 disposition between mouse and human BBB models. In conclusion, current study demonstrates the important role of BBB in the clearance of Aβ from the brain. Moreover, it provides insight into the differences between mouse and human BBB with regards to Aβ clearance and offer, for the first time, a mathematical model that describes Aβ clearance across BBB.


Free Radical Biology and Medicine | 2008

Protective effect of Pycnogenol in human neuroblastoma SH-SY5Y cells following acrolein-induced cytotoxicity.

Mubeen A. Ansari; Jeffrey N. Keller; Stephen W. Scheff

Oxidative stress is one of the hypotheses involved in the etiology of Alzheimers disease (AD). Considerable attention has been focused on increasing the intracellular glutathione (GSH) levels in many neurodegenerative diseases, including AD. Pycnogenol (PYC) has antioxidant properties and stabilizes intracellular antioxidant defense systems including glutathione levels. The present study investigated the protective effects of PYC on acrolein-induced oxidative cell toxicity in cultured SH-SY5Y neuroblastoma cells. Decreased cell survival in SH-SY5Y cultures treated with acrolein correlated with oxidative stress, increased NADPH oxidase activity, free radical production, protein oxidation/nitration (protein carbonyl, 3-nitrotyrosine), and lipid peroxidation (4-hydroxy-2-nonenal). Pretreatment with PYC significantly attenuated acrolein-induced cytotoxicity, protein damage, lipid peroxidation, and cell death. A dose-response study suggested that PYC showed protective effects against acrolein toxicity by modulating oxidative stress and increasing GSH. These findings provide support that PYC may provide a promising approach for the treatment of oxidative stress-related neurodegenerative diseases such as AD.


ACS Chemical Neuroscience | 2015

Oleocanthal enhances amyloid-β clearance from the brains of TgSwDI mice and in vitro across a human blood-brain barrier model.

Hisham Qosa; Yazan S. Batarseh; Mohamed M. Mohyeldin; Khalid A. El Sayed; Jeffrey N. Keller; Amal Kaddoumi

Numerous clinical and preclinical studies have suggested several health promoting effects for the dietary consumption of extra-virgin olive oil (EVOO) that could protect and decrease the risk of developing Alzheimer’s disease (AD). Moreover, recent studies have linked this protective effect to oleocanthal, a phenolic secoiridoid component of EVOO. This protective effect of oleocanthal against AD has been related to its ability to prevent amyloid-β (Aβ) and tau aggregation in vitro, and enhance Aβ clearance from the brains of wild type mice in vivo; however, its effect in a mouse model of AD is not known. In the current study, we investigated the effect of oleocanthal on pathological hallmarks of AD in TgSwDI, an animal model of AD. Mice treatment for 4 weeks with oleocanthal significantly decreased amyloid load in the hippocampal parenchyma and microvessels. This reduction was associated with enhanced cerebral clearance of Aβ across the blood-brain barrier (BBB). Further mechanistic studies demonstrated oleocanthal to increase the expression of important amyloid clearance proteins at the BBB including P-glycoprotein and LRP1, and to activate the ApoE-dependent amyloid clearance pathway in the mice brains. The anti-inflammatory effect of oleocanthal in the brains of these mice was also obvious where it was able to reduce astrocytes activation and IL-1β levels. Finally, we could recapitulate the observed protective effect of oleocanthal in an in vitro human-based model, which could argue against species difference in response to oleocanthal. In conclusion, findings from in vivo and in vitro studies provide further support for the protective effect of oleocanthal against the progression of AD.


Biochimica et Biophysica Acta | 2014

Mixed oligomers and monomeric Amyloid-β disrupts endothelial cells integrity and reduces monomeric amyloid-β transport across hCMEC/D3 cell line as an in vitro blood-brain barrier model

Hisham Qosa; Harry LeVine; Jeffrey N. Keller; Amal Kaddoumi

Senile amyloid plaques are one of the diagnostic hallmarks of Alzheimers disease (AD). However, the severity of clinical symptoms of AD is weakly correlated with the plaque load. AD symptoms severity is reported to be more strongly correlated with the level of soluble amyloid-β (Aβ) assemblies. Formation of soluble Aβ assemblies is stimulated by monomeric Aβ accumulation in the brain, which has been related to its faulty cerebral clearance. Studies tend to focus on the neurotoxicity of specific Aβ species. There are relatively few studies investigating toxic effects of Aβ on the endothelial cells of the blood-brain barrier (BBB). We hypothesized that a soluble Aβ pool more closely resembling the in vivo situation composed of a mixture of Aβ40 monomer and Aβ42 oligomer would exert higher toxicity against hCMEC/D3 cells as an in vitro BBB model than either component alone. We observed that, in addition to a disruptive effect on the endothelial cells integrity due to enhancement of the paracellular permeability of the hCMEC/D3 monolayer, the Aβ mixture significantly decreased monomeric Aβ transport across the cell culture model. Consistent with its effect on Aβ transport, Aβ mixture treatment for 24h resulted in LRP1 down-regulation and RAGE up-regulation in hCMEC/D3 cells. The individual Aβ species separately failed to alter Aβ clearance or the cell-based BBB model integrity. Our study offers, for the first time, evidence that a mixture of soluble Aβ species, at nanomolar concentrations, disrupts endothelial cells integrity and its own transport across an in vitro model of the BBB.


Journal of Nutritional Biochemistry | 2015

Extra-virgin olive oil attenuates amyloid-β and tau pathologies in the brains of TgSwDI mice.

Hisham Qosa; Loqman A. Mohamed; Yazan S. Batarseh; Saeed Alqahtani; Baher A. Ibrahim; Harry LeVine; Jeffrey N. Keller; Amal Kaddoumi

Extra-virgin olive oil (EVOO) is one of the main elements of Mediterranean diet. Several studies have suggested that EVOO has several health promoting effects that could protect from and decrease the risk of Alzheimers disease (AD). In this study, we investigated the effect of consumption of EVOO-enriched diet on amyloid- and tau-related pathological alterations that are associated with the progression of AD and cerebral amyloid angiopathy (CAA) in TgSwDI mice. Feeding mice with EVOO-enriched diet for 6months, beginning at an age before amyloid-β (Aβ) accumulation starts, has significantly reduced total Aβ and tau brain levels with a significant improvement in mouse cognitive behavior. This reduction in brain Aβ was explained by the enhanced Aβ clearance pathways and reduced brain production of Aβ via modulation of amyloid-β precursor protein processing. On the other hand, although feeding mice with EVOO-enriched diet for 3months, beginning at an age after Aβ accumulation starts, showed improved clearance across the blood-brain barrier and significant reduction in Aβ levels, it did not affect tau levels or improve cognitive functions of TgSwDI mouse. Collectively, results of this study suggest that the long-term consumption of EVOO-containing diet starting at early age provides a protective effect against AD and its related disorder CAA.


Biochimica et Biophysica Acta | 2016

Role of P-glycoprotein in mediating rivastigmine effect on amyloid-β brain load and related pathology in Alzheimer's disease mouse model

Loqman A. Mohamed; Jeffrey N. Keller; Amal Kaddoumi

Recently, we showed that rivastigmine decreased amyloid-β (Aβ) brain load in aged rats by enhancing its clearance across the blood-brain barrier (BBB) via upregulation of P-glycoprotein (P-gp) and low-density lipoprotein receptor-related protein 1 (LRP1). Here, we extend our previous work to clarify P-gp role in mediating rivastigmine effect on Aβ brain levels and neuroprotection in a mouse model of Alzheimers disease (AD) that expresses different levels of P-gp. APPSWE mice were bred with mdr1a/b knockout mice to produce littermates that were divided into three groups; APP(+)/mdr1(+/+), APP(+)/mdr1(+/-) and APP(+)/mdr1(-/-). Animals received rivastigmine treatment (0.3mg/kg/day) or vehicle for 8weeks using Alzet osmotic mini-pumps. ELISA analysis of brain homogenates for Aβ showed rivastigmine treatment to significantly decrease Aβ brain load in APP(+)/mdr1(+/+) by 25% and in APP(+)/mdr1(+/-) mice by 21% compared to their vehicle treated littermates, but not in APP(+)/mdr1(-/-) mice. In addition, rivastigmine reduced GFAP immunostaining of astrocytes by 50% and IL-1β brain level by 43% in APP(+)/mdr1(+/+) mice, however its effect was less pronounced in P-gp knockout mice. Moreover, rivastigmine demonstrated a P-gp expression dependent neuroprotective effect that was highest in APP(+)/mdr1(+/+)>APP(+)/mdr1(+/-)>APP(+)/mdr1(-/-) as determined by expression of synaptic markers PSD-95 and SNAP-25 using Western blot analysis. Collectively, our results suggest that P-gp plays important role in mediating rivastigmine non-cholinergic beneficial effects, including Aβ brain load reduction, neuroprotective and anti-inflammatory effects in the AD mouse models.


Biochimica et Biophysica Acta | 2013

Prolonged diet induced obesity has minimal effects towards brain pathology in mouse model of cerebral amyloid angiopathy: Implications for studying obesity–brain interactions in mice

Le Zhang; Kalavathi Dasuri; Sun-Ok Fernandez-Kim; Annadora J. Bruce-Keller; Linnea R. Freeman; Jennifer K. Pepping; Tina L. Beckett; M. Paul Murphy; Jeffrey N. Keller

Cerebral amyloid angiopathy (CAA) occurs in nearly every individual with Alzheimers disease (AD) and Downs syndrome, and is the second largest cause of intracerebral hemorrhage. Mouse models of CAA have demonstrated evidence for increased gliosis contributing to CAA pathology. Nearly two thirds of Americans are overweight or obese, with little known about the effects of obesity on the brain, although increasingly the vasculature appears to be a principle target of obesity effects on the brain. In the current study we describe for the first time whether diet induced obesity (DIO) modulates glial reactivity, amyloid levels, and inflammatory signaling in a mouse model of CAA. In these studies we identify surprisingly that DIO does not significantly increase Aβ levels, astrocyte (GFAP) or microglial (IBA-1) gliosis in the CAA mice. However, within the hippocampal gyri a localized increase in reactive microglia were increased in the CA1 and stratum oriens relative to CAA mice on a control diet. DIO was observed to selectively increase IL-6 in CAA mice, with IL-1β and TNF-α not increased in CAA mice in response to DIO. Taken together, these data show that prolonged DIO has only modest effects towards Aβ in a mouse model of CAA, but appears to elevate some localized microglial reactivity within the hippocampal gyri and selective markers of inflammatory signaling. These data are consistent with the majority of the existing literature in other models of Aβ pathology, which surprisingly show a mixed profile of DIO effects towards pathological processes in mouse models of neurodegenerative disease. The importance for considering the potential impact of ceiling effects in pathology within mouse models of Aβ pathogenesis, and the current experimental limitations for DIO in mice to fully replicate metabolic dysfunction present in human obesity, are discussed. This article is part of a Special Issue entitled: Animal Models of Disease.


Journal of Clinical and Experimental Neuropsychology | 2018

Understanding heterogeneity in older adults: Latent growth curve modeling of cognitive functioning

Rebecca K. MacAulay; Matthew Calamia; Alex S. Cohen; Katrina Daigle; Heather Foil; Robert Brouillette; Annadora J. Bruce-Keller; Jeffrey N. Keller

ABSTRACT Background: Clarifying relationships between specific neurocognitive functions in cognitively intact older adults can improve our understanding of mechanisms involved in cognitive decline, which may allow identification of new opportunities for intervention and earlier detection of those at increased risk of dementia. Method: The present study employed latent growth curve modeling to longitudinally examine the relationship between executive attention/processing speed, episodic memory, language, and working memory functioning utilizing the neuropsychological test battery from the National Alzheimer’s Disease Coordinating Center. A total of 691 relatively healthy older adults (Mage = 69.07, SD = 6.49) were assessed at baseline, and 553 individuals completed three visits spanning a two-year period. Results: Better cognitive performance was concomitantly associated with better functioning across domains. Subtle declines in executive attention/processing speed processes were found, while, on average, memory and language performance improved with repeated testing. Lower executive attention/processing speed performance at baseline predicted less incremental growth rate in memory. In turn, higher initial memory functioning was associated with incremental improvements in language performance. Conclusions: These results are consistent with the notion that intact executive function and attention processes are important to preserving memory functioning with advanced age, but are also the functions most susceptible to decline with age. These findings also provide further insight into the critical role of practice effects in clinical assessment practice and have implications for pharmaceutical trials. Practice effects should be routinely considered as they may give the appearance of retention of function within the cognitive domains considered to be a hallmark of Alzheimer’s disease pathology.


2016 32nd Southern Biomedical Engineering Conference (SBEC) | 2016

Development of an in-vitro High-Throughput Screening Assay for the Identification of Modulators of the Blood-Brain Barrier Endothelium Integrity

Sweilem B. Al Rihani; Hisham Qosa; Loqman A. Mohamed; Yazan S. Batarseh; Jeffrey N. Keller; Amal Kaddoumi

The blood-brain barrier (BBB) controls the content of brain interstitial fluid due to the presence of high-resistance tight junction proteins between the endothelial cells of brain capillaries. Several models using different cell-lines were developed to study the BBB biology, these models were complex and their use for high-throughput screening (HTS) has proven to be challenging. Therefore, we developed an in-vitro BBB model that is practical and reliable for HTS. The mouse brain endothelial cells (bEnd3) grown on 96-well plate inserts were upgraded and optimized for HTS assay. Using Lucifer Yellow (LY) permeation assay, 3 different compounds libraries that include 3000 compounds were screened for hits that have the potential to modulate the BBB model integrity. To evaluate the model ability to identify compounds that increase LY permeation, mannitol was used as a positive control for disruptors. The model performance in this assay was high with Z factor above 0.5 and high S/N and S/B ratios. Alternatively, hydrocortisone was used as a positive control for compounds that enhance the barrier function as it is known to improve endothelium tightness. The Z factor determined with hydrocortisone was 0.3 with lower S/N and S/B ratios compared to mannitol .The primary screen has identified several hundred of modulators. The secondary screen could identify 13 compounds as potent enhancers of the monolayer integrity with EC50 values less than 10 μM. In conclusion, an HTS-BBB model was developed and used for compounds screening to identify compelling hits for further evaluation for their effect on the BBB.

Collaboration


Dive into the Jeffrey N. Keller's collaboration.

Top Co-Authors

Avatar

Amal Kaddoumi

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Hisham Qosa

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Yazan S. Batarseh

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Annadora J. Bruce-Keller

Pennington Biomedical Research Center

View shared research outputs
Top Co-Authors

Avatar

Loqman A. Mohamed

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alex S. Cohen

Louisiana State University

View shared research outputs
Top Co-Authors

Avatar

Anthony Giordano

Louisiana State University

View shared research outputs
Top Co-Authors

Avatar

Ashley Depaula

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge