Network


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

Hotspot


Dive into the research topics where James Keblesh is active.

Publication


Featured researches published by James Keblesh.


Molecular and Cellular Neuroscience | 2007

Egr3, a synaptic activity regulated transcription factor that is essential for learning and memory

Lin Li; Sung Hwan Yun; James Keblesh; Barbara L. Trommer; Huangui Xiong; Jelena Radulovic; Warren G. Tourtellotte

Learning and memory depend upon poorly defined synaptic and intracellular modifications that occur in activated neurons. Mitogen activated protein kinase-extracellular regulated kinase (MAPK-ERK) signaling and de novo protein synthesis are essential aspects of enduring memory formation, but the precise effector molecules of MAPK-ERK signaling in neurons are not well defined. Early growth response (Egr) transcriptional regulators are examples of MAPK-ERK regulated genes and Egr1 (zif268) has been widely recognized as essential for some aspects of learning and memory. Here we show that Egr3, a transcriptional regulator closely related to Egr1, is essential for normal hippocampal long-term potentiation (LTP) and for hippocampal and amygdala dependent learning and memory. In the absence of Egr3, the defects in learning and memory appear to be independent of Egr1 since Egr1 protein levels are not altered in amygdala, hippocampus or cortex. Moreover, unlike Egr1-deficient mice which have impairments in late phase hippocampal LTP and consolidation of some forms of long-term hippocampus- and amygdala-dependent memory, Egr3-deficient mice have profound defects in early- and late-phase hippocampal LTP, as well as short-term and long-term hippocampus- and amygdala-dependent learning and memory. Thus, Egr3 has an essential role in learning and memory processing that appears to be partly distinct from the role of Egr1.


Free Radical Biology and Medicine | 2010

Acetyl-L-carnitine protects neuronal function from alcohol-induced oxidative damage in the brain.

Travis J. Rump; P. M. Abdul Muneer; Adam M. Szlachetka; Allyson Lamb; Catherine Haorei; Saleena Alikunju; Huangui Xiong; James Keblesh; Jianuo Liu; Matthew C. Zimmerman; Jocelyn Jones; Terrence M. Donohue; Yuri Persidsky; James Haorah

The studies presented here demonstrate the protective effect of acetyl-L-carnitine (ALC) against alcohol-induced oxidative neuroinflammation, neuronal degeneration, and impaired neurotransmission. Our findings reveal the cellular and biochemical mechanisms of alcohol-induced oxidative damage in various types of brain cells. Chronic ethanol administration to mice caused an increase in inducible nitric oxide synthase (iNOS) and 3-nitrotyrosine adduct formation in frontal cortical neurons but not in astrocytes from brains of these animals. Interestingly, alcohol administration caused a rather selective activation of NADPH oxidase (NOX), which, in turn, enhanced levels of reactive oxygen species (ROS) and 4-hydroxynonenal, but these were predominantly localized in astrocytes and microglia. Oxidative damage in glial cells was accompanied by their pronounced activation (astrogliosis) and coincident neuronal loss, suggesting that inflammation in glial cells caused neuronal degeneration. Immunohistochemistry studies indicated that alcohol consumption induced different oxidative mediators in different brain cell types. Thus, nitric oxide was mostly detected in iNOS-expressing neurons, whereas ROS were predominantly generated in NOX-expressing glial cells after alcohol ingestion. Assessment of neuronal activity in ex vivo frontal cortical brain tissue slices from ethanol-fed mice showed a reduction in long-term potentiation synaptic transmission compared with slices from controls. Coadministration of ALC with alcohol showed a significant reduction in oxidative damage and neuronal loss and a restoration of synaptic neurotransmission in this brain region, suggesting that ALC protects brain cells from ethanol-induced oxidative injury. These findings suggest the potential clinical utility of ALC as a neuroprotective agent that prevents alcohol-induced brain damage and development of neurological disorders.


PLOS ONE | 2011

HIV-1gp120 Induces Neuronal Apoptosis through Enhancement of 4-Aminopyridine-Senstive Outward K+ Currents

Lina Chen; Jianuo Liu; Changshui Xu; James Keblesh; Wei-Jin Zang; Huangui Xiong

Human immunodeficiency virus type 1 (HIV-1)-associated dementia (HAD) usually occurs late in the course of HIV-1 infection and the mechanisms underlying HAD pathogenesis are not well understood. Accumulating evidence indicates that neuronal voltage-gated potassium (Kv) channels play an important role in memory processes and acquired neuronal channelopathies in HAD. To examine whether Kv channels are involved in HIV-1-associated neuronal injury, we studied the effects of HIV-1 glycoprotein 120 (gp120) on outward K+ currents in rat cortical neuronal cultures using whole-cell patch techniques. Exposure of cortical neurons to gp120 produced a dose-dependent enhancement of A-type transient outward K+ currents (IA). The gp120-induced increase of IA was attenuated by T140, a specific antagonist for chemokine receptor CXCR4, suggesting gp120 enhancement of neuronal IA via CXCR4. Pretreatment of neuronal cultures with a protein kinase C (PKC) inhibitor, GF109203X, inhibited the gp120-induced increase of IA. Biological significance of gp120 enhancement of IA was demonstrated by experimental results showing that gp120-induced neuronal apoptosis, as detected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and caspase-3 staining, was attenuated by either an IA blocker 4-aminopyridine or a specific CXCR4 antagonist T140. Taken together, these results suggest that gp120 may induce caspase-3 dependent neuronal apoptosis by enhancing IA via CXCR4-PKC signaling.


Journal of Neuroimmune Pharmacology | 2009

4-aminopyridine improves spatial memory in a murine model of HIV-1 encephalitis

James Keblesh; Huanyu Dou; Howard E. Gendelman; Huangui Xiong

HIV-1-associated neurocognitive disorders (HAND) remains a significant source of morbidity in the era of wide spread use of highly active antiretroviral therapy. Disease is precipitated by low levels of viral growth and glial immune activation within the central nervous system. Blood borne macrophage and microglia affect a proinflammatory response and release viral proteins that affects neuronal viability and leads to death of nerve cells. Increasing evidence supports the notion that HAND is functional channelopathy, but proof of this concept remains incomplete. Based on their role in learning and memory processes, we now posit that voltage-gated potassium (Kv) channels could be a functional substrate for disease. This was tested in the severe combined immunodeficient (SCID) mouse model of HIV-1 encephalitis (HIVE) by examining whether the Kv channel blocker, 4-aminopyridine (4-AP), could affect behavioral, electrophysiological, and morphological measures of learning and memory. HIVE SCID mice showed impaired spatial memory in radial arm water maze tests. Electrophysiology studies revealed a reduction of long-term potentiation (LTP) in the CA1 region of the hippocampus. Importantly, systemic administration of 4-AP blocked HIV-1-associated reduction of LTP and improved animal performance in the radial arm water maze. These results support the importance of Kv channel dysfunction in disease but, more importantly, provide a potential target for adjunctive therapies for HAND.


PLOS ONE | 2013

HIV-1 Tat Protein Increases Microglial Outward K+ Current and Resultant Neurotoxic Activity

Jianuo Liu; Peng Xu; Cory Collins; Han Liu; Jingdong Zhang; James Keblesh; Huangui Xiong

Microglia plays a crucial role in the pathogenesis of HIV-1-associated neurocognitive disorders. Increasing evidence indicates the voltage-gated potassium (Kv) channels are involved in the regulation of microglia function, prompting us to hypothesize Kv channels may also be involved in microglia-mediated neurotoxic activity in HIV-1-infected brain. To test this hypothesis, we investigated the involvement of Kv channels in the response of microglia to HIV-1 Tat protein. Treatment of rat microglia with HIV-1 Tat protein (200 ng/ml) resulted in pro-inflammatory microglial activation, as indicated by increases in TNF-α, IL-1β, reactive oxygen species, and nitric oxide, which were accompanied by enhanced outward K+ current and Kv1.3 channel expression. Suppression of microglial Kv1.3 channel activity, either with Kv1.3 channel blockers Margatoxin, 5-(4-Phenoxybutoxy)psoralen, or broad-spectrum K+ channel blocker 4-Aminopyridine, or by knockdown of Kv1.3 expression via transfection of microglia with Kv1.3 siRNA, was found to abrogate the neurotoxic activity of microglia resulting from HIV-1 Tat exposure. Furthermore, HIV-1 Tat-induced neuronal apoptosis was attenuated with the application of supernatant collected from K+ channel blocker-treated microglia. Lastly, the intracellular signaling pathways associated with Kv1.3 were investigated and enhancement of microglial Kv1.3 was found to correspond with an increase in Erk1/2 mitogen-activated protein kinase activation. These data suggest targeting microglial Kv1.3 channels may be a potential new avenue of therapy for inflammation-mediated neurological disorders.


Journal of Neuroimmune Pharmacology | 2009

Voltage-gated Potassium Channels in Human Immunodeficiency Virus Type-1 (HIV-1)-associated Neurocognitive Disorders

James Keblesh; Dehui Hu; Huangui Xiong

Human immunodeficiency virus type-1 (HIV-1)-associated dementia (HAD), a severe form of HIV-associated neurocognitive disorders (HAND), describes the cognitive impairments and behavioral disturbances which afflict many HIV-infected individuals. Although the precise mechanism leading to HAD is incompletely understood, it is commonly accepted its progression involves a critical mass of infected and activated mononuclear phagocytes (brain perivascular macrophages and microglia) releasing immune and viral products in the brain. These cellular and viral products induce neuronal dysfunction and injury via various signaling pathways. Emerging evidence indicates voltage-gated potassium (Kv) channels, key regulators of cell excitability and animal behavior (learning and memory), are involved in the pathogenesis of HAD/HAND. Here we survey the literature and find that HAD-related alterations in cellular and viral products can increase neuronal Kv channel activity, leading to neuronal dysfunction and cognitive deficits. Thus, neuronal Kv channels may be a new target in the effort to develop therapies for HAD and perhaps other inflammatory neurodegenerative disorders.


European Journal of Neuroscience | 2010

Involvement of the 4-aminopyridine-sensitive transient A-type K+ current in macrophage-induced neuronal injury

Dehui Hu; Jianuo Liu; James Keblesh; Huangui Xiong

Through their capacity to secrete, upon activation, a variety of bioactive molecules, brain macrophages (and resident microglia) play an important role in brain immune and inflammatory responses. To test our hypothesis that activated macrophages induce neuronal injury by enhancing neuronal outward K+ current, we studied the effects of lipopolysaccharide (LPS)‐stimulated human monocyte‐derived macrophage (MDM) on neuronal transient A‐type K+ current (IA) and resultant neuronal injury in primary rat hippocampal neuronal cultures. Bath application of LPS‐stimulated MDM‐conditioned media (MCM+) enhanced neuronal IA in a concentration‐dependent manner. Non‐stimulated MCM (MCM‐) failed to alter IA. The enhancement of neuronal IA was recapitulated in neurons co‐cultured with macrophages. The link of MCM(+)‐induced enhancement of IA to MCM(+)‐associated neuronal injury, as detected by propidium iodide and 4″,6‐diamidino‐2‐phenylindol staining (DAPI) and 3‐(4,5‐dimethylthiazol‐2‐yl)‐2,5‐diphenyl tetrazolium bromide (MTT) assay, was demonstrated by experimental results showing that addition of IA blocker 4‐aminopyridine to the cultures protected hippocampal neurons from MCM(+)‐induced neuronal injury. Further investigation revealed that glutamate was involved in MCM(+)‐induced enhancement of neuronal IA. These results suggest that during brain inflammation macrophages (and microglia) might mediate neuronal injury via enhancement of neuronal IA, and that neuronal Kv channel might be a potential target for the development of therapeutic strategies for some neurodegenerative disorders by which immune and inflammatory responses are believed to be involved in the pathogenesis.


Journal of Neuroimmune Pharmacology | 2007

Voltage-gated potassium channel modulation of neurotoxic activity in human immunodeficiency virus type-1(HIV-1)-infected macrophages.

Elizabeth Irvine; James Keblesh; Jianuo Liu; Huangui Xiong

Macrophages play an important role in brain immune and inflammatory responses. They are also critical cells in mediating the pathology of neurodegenerative disorders such as HIV-associated dementia. This is largely through their capacity to secrete a variety of bioactive molecules such as cytokines, leading to neuronal dysfunction and/or death. Accumulating evidence indicates that voltage-gated potassium (Kv) channels play a pivotal role in the modulation of macrophage proliferation, activation, and secretion. Blockade of Kv channels by specific antagonists decreases macrophage cytokine production and ameliorates macrophage-associated neuronal injury. These results suggest that Kv channels might become a potential target for the development of new therapeutic strategies for chronic inflammatory diseases.


Retrovirology: Research and Treatment | 2008

Pathogenesis of Human Immunodeficiency Virus Type-1 (HIV-1)-Associated Dementia: Role of Voltage-Gated Potassium Channels

James Keblesh; Benjamin C. Reiner; Jianuo Liu; Huangui Xiong

HIV-1-associated dementia (HAD) describes the cognitive impairments and behavioral disturbances which afflict many HIV-infected individuals. Although the incidence of HAD has decreased significantly in the era of HAART, it remains a significant complication of HIV-1 infection as patients with acquired immune deficient syndrome (AIDS) live longer, antiretroviral drugs remain unable to effectively cross the blood-brain barrier (BBB), and HIV-1 resistance grows due to viral strain mutation. Although the precise mechanism leading to HAD is incompletely understood, it is commonly accepted its progression involves a critical mass of infected and activated mononuclear phagocytes (MP; brain perivascular macrophages and microglia) releasing immune and viral products in brain. These cellular and viral products induce neuronal dysfunction and injury via various signaling pathways. Emerging evidence indicates that voltage-gated potassium (K(v)) channels, key regulators of cell excitability and animal behavior (learning and memory), are involved in the pathogenesis of HAD/HAND. Here we survey the literature and find HAD related alterations in cellular and viral products can alter MP and neuronal K(v) channel activity, leading to MP and neuronal dysfunction and cognitive deficits. Thus, MP and neuronal K(v) channels may be a new target in the effort to develop therapies for HAD and perhaps other inflammatory neurodegenerative disorders.


International journal of physiology, pathophysiology and pharmacology | 2009

Methamphetamine abuse, HIV infection, and neurotoxicity

Benjamin C. Reiner; James Keblesh; Huangui Xiong

Collaboration


Dive into the James Keblesh's collaboration.

Top Co-Authors

Avatar

Huangui Xiong

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jianuo Liu

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Benjamin C. Reiner

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dehui Hu

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Adam M. Szlachetka

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Allyson Lamb

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Catherine Haorei

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Changshui Xu

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Cory Collins

University of Nebraska Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge