Network


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

Hotspot


Dive into the research topics where Avtar K. Singh is active.

Publication


Featured researches published by Avtar K. Singh.


Biochemical and Biophysical Research Communications | 2010

Involvement of AMP-activated-protein-kinase (AMPK) in neuronal amyloidogenesis

Je-Seong Won; Yeong-Bin Im; Jinsu Kim; Avtar K. Singh; Inderjit Singh

AMP-activated-protein-kinase (AMPK) is a key sensor and regulator of cellular and whole-body energy metabolism and plays a key role in regulation of lipid metabolism. Since lipid metabolism has been implicated in neuronal amyloid-beta (Abeta) homeostasis and onset of Alzheimers disease, we investigated the involvement of AMPK in neuronal lipid metabolism and Abeta production. We observed in cultured rat cortical neurons that Abeta production was significantly reduced when the neurons were stimulated with AMPK activator, 5-aminoimidazole-4-carboxamide-1-d-ribofuranoside (AICAR), but increased when AMPKalpha2 was knocked out, thus indicating the role of AMPK in amyloidogenesis. Although the detailed mechanisms by which AMPK regulates Abeta generation is not well understood, AMPK-mediated alterations in cholesterol and sphingomyelin homeostasis and in turn the altered distribution of Abeta precursor-protein (APP) in cholesterol and sphingomyelin rich membrane lipid rafts participate in Abeta generation. Taken together, this is the first report on the role of AMPK in regulation of neuronal amyloidogenesis.


Journal of Neurochemistry | 2008

Lovastatin inhibits amyloid precursor protein (APP) β-cleavage through reduction of APP distribution in Lubrol WX extractable low density lipid rafts

Je-Seong Won; Yeong-Bin Im; Mushfiquddin Khan; Miguel A. Contreras; Avtar K. Singh; Inderjit Singh

Previous studies have described that statins (inhibitors of cholesterol and isoprenoid biosynthesis) inhibit the output of amyloid‐β (Aβ) in the animal model and thus decrease risk of Alzheimer’s disease. However, their action mechanism(s) in Aβ precursor protein (APP) processing and Aβ generation is not fully understood. In this study, we report that lovastatin treatment reduced Aβ output in cultured hippocampal neurons as a result of reduced APP levels and β‐secretase activities in low density Lubrol WX (non‐ionic detergent) extractable lipid rafts (LDLR). Rather than altering cholesterol levels in lipid raft fractions and thus disrupting lipid raft structure, lovastatin decreased Aβ generation through down‐regulating geranylgeranyl‐pyrophosphate dependent endocytosis pathway. The inhibition of APP endocytosis by treatment with lovastatin and reduction of APP levels in LDLR fractions by treatment with phenylarsine oxide (a general endocytosis inhibitor) support the involvement of APP endocytosis in APP distribution in LDLR fractions and subsequent APP β‐cleavage. Moreover, lovastatin‐mediated down‐regulation of endocytosis regulators, such as early endosomal antigen 1, dynamin‐1, and phosphatidylinositol 3‐kinase activity, indicates that lovastatin modulates APP endocytosis possibly through its pleiotropic effects on endocytic regulators. Collectively, these data report that lovastatin mediates inhibition of LDLR distribution and β‐cleavage of APP in a geranylgeranyl‐pyrophosphate and endocytosis‐dependent manner.


Journal of Neurochemistry | 2007

Lactosylceramide: a lipid second messenger in neuroinflammatory disease

Je-Seong Won; Avtar K. Singh; Inderjit Singh

Inflammatory disease plays a critical role in the pathogenesis of many neurological disorders. Astrogliosis and induction of pro‐inflammatory mediators such as chemokines, cytokines and inducible nitric oxide synthase (iNOS) are the ‘hallmarks’ of inflammatory disease. Increased activity of lactosylceramide (LacCer) synthase and increased synthesis of LacCer during glial proliferation, and induction of pro‐inflammatory cytokines and iNOS suggests a role for LacCer in these cellular signaling pathways. Studies using complementary techniques of inhibitors and antisense reported that inhibition of LacCer synthesis inhibits glial proliferation, as well as the induction of pro‐inflammatory mediators (cytokines and iNOS). This inhibition was bypassed by exogenous LacCer, but not by other related lipids (e.g. glucosylceramide, galactocerebroside, GD1, GM1), indicating a role for LacCer in inflammatory signaling pathways. Furthermore, inhibition of glial proliferation and induction of inflammatory mediators by antisense to Ras GTPase, PI3Kinase and inhibitors of mitogen‐activated protein kinase indicate the participation of the phosphoinositide 3‐kinase (PI3Kinas)/Ras/mitogen‐activated protein kinase/nuclear factor‐κB (NF‐κB) signaling pathways in LacCer‐mediated inflammatory events thus exposing additional targets for therapeutics for inflammatory disease conditions.


Glia | 2005

Involvement of phospholipase A2 and lipoxygenase in lipopolysaccharide-induced inducible nitric oxide synthase expression in glial cells

Je-Seong Won; Yeong-Bin Im; Mushfiquddin Khan; Avtar K. Singh; Inderjit Singh

The present study underlines the importance of phospholipase A2 (PLA2)‐ and lipoxygenase (LO)‐mediated signaling processes in the regulation of inducible nitric oxide synthase (iNOS) gene expression. In glial cells, lipopolysaccharide (LPS) induced the activities of PLA2 (calcium‐independent PLA2; iPLA2 and cytosolic PLA2; cPLA2) as well as gene expression of iNOS. The inhibition of cPLA2 by methyl arachidonyl fluorophosphates (MAFP) or antisense oligomer against cPLA2 and inhibition of iPLA2 by bromoenol lactone reduced the LPS‐induced iNOS gene expression and NFκB activation. In addition, the inhibition of LO by nordihydroguaiaretic acid (NDGA; general LO inhibitor) or MK886 (5‐LO inhibitor), but not baicalein (12‐LO inhibitor), completely abrogated the LPS‐induced iNOS expression. Because NDGA could abrogate the LPS‐induced activation of NFκB, while MK886 had no effect on it, LO‐mediated inhibition of iNOS gene induction by LPS may involve an NFκB‐dependent or ‐independent (by 5‐LO) pathway. In contrast to LO, however, the cyclooxygenase (COX) may not be involved in the regulation of LPS‐mediated induction of iNOS gene because COX inhibition by indomethacin (general COX inhibitor), SC560 (COX‐1 inhibitor), and NS398 (COX‐2 inhibitor) affected neither the LPS‐induced iNOS expression nor activation of NFκB. These results indicate a role for cPLA2 and iPLA2 in LPS‐mediated iNOS gene induction in glial cells and the involvement of LO in these reactions.


Journal of Alzheimer's Disease | 2013

Protective Role of S-Nitrosoglutathione (GSNO) Against Cognitive Impairment in Rat Model of Chronic Cerebral Hypoperfusion

Je-Seong Won; Jinsu Kim; Balasubramaniam Annamalai; Anandakumar Shunmugavel; Inderjit Singh; Avtar K. Singh

Chronic cerebral hypoperfusion (CCH), featuring in most of the Alzheimers disease spectrum, plays a detrimental role in brain amyloid-β (Aβ) homeostasis, cerebrovascular morbidity, and cognitive decline; therefore, early management of cerebrovascular pathology is considered to be important for intervention in the impending cognitive decline. S-nitrosoglutathione (GSNO) is an endogenous nitric oxide carrier modulating endothelial function, inflammation, and neurotransmission. Therefore, the effect of GSNO treatment on CCH-associated neurocognitive pathologies was determined in vivo by using rats with permanent bilateral common carotid artery occlusion (BCCAO), a rat model of chronic cerebral hypoperfusion. We observed that rats subjected to permanent BCCAO showed a significant decrease in learning/memory performance and increases in brain levels of Aβ and vascular inflammatory markers. GSNO treatment (50 μg/kg/day for 2 months) significantly improved learning and memory performance of BCCAO rats and reduced the Aβ levels and ICAM-1/VCAM-1 expression in the brain. Further, in in vitro cell culture studies, GSNO treatment also decreased the cytokine-induced proinflammatory responses, such as activations of NFκB and STAT3 and expression of ICAM-1 and VCAM-1 in endothelial cells. In addition, GSNO treatment increased the endothelial and microglial Aβ uptake. Additionally, GSNO treatment inhibited the β-secretase activity in primary rat neuron cell culture, thus reducing secretion of Aβ, suggesting GSNO mediated mechanisms in anti-inflammatory and anti-amyloidogenic activities. Taken together, these data document that systemic GSNO treatment is beneficial for improvement of cognitive decline under the conditions of chronic cerebral hypoperfusion and suggests a potential therapeutic use of GSNO for cerebral hypoperfusion associated mild cognitive impairment in Alzheimers disease.


Brain Research | 2013

Role of endogenous psychosine accumulation in oligodendrocyte differentiation and survival: implication for Krabbe disease.

Je-Seong Won; Jinsu Kim; Manjeet K. Paintlia; Inderjit Singh; Avtar K. Singh

Krabbe disease is a lethal, demyelinating condition caused by genetic deficiency of galactocerebrosidase (GALC) and resultant accumulation of its cytotoxic substrate, psychosine (galactosylsphingosine), primarily in oligodendrocytes (OLs). Psychosine is generated by galactosylation of sphingosine by UDP-galactose:ceramide galactosyltransferase (CGT), a galactosylceramide synthesizing enzyme which is primarily expressed in OLs. The expression of CGT and the synthesis of galactosyl-sphingolipids are associated with the terminal differentiation of OL, but little is known about the participation of endogenous psychosine accumulation in OL differentiation under GALC deficient conditions. In this study, we report that accumulation of endogenous psychosine under GALC deficient Krabbe conditions impedes OL differentiation process both by decreasing the expression of myelin lipids and protein and by inducing the cell death of maturating OLs. The psychosine pathology under GALC deficient conditions involves participation of secretory phospholipase A2 (sPLA2) activation and increase in its metabolites, as evidenced by attenuation of psychosine-induced pathology by treatment with pharmacological inhibitor of sPLA2 7,7-dimethyleicosadienoic acid (DEDA). These observations suggest for potential therapeutic efficacy of sPLA2 inhibitor in Krabbe disease.


Biochemical and Biophysical Research Communications | 2015

Role of S-nitrosoglutathione mediated mechanisms in tau hyper-phosphorylation

Balasubramaniam Annamalai; Je-Seong Won; Seungho Choi; Inderjit Singh; Avtar K. Singh

Hyperphosphorylation and polymerization of microtubule-associated protein tau into paired helical filaments (PHFs) is one of the hallmarks of Alzheimers disease (AD). Here we report that neuronal tau hyperphosphorylation under AD conditions is regulated by S-nitrosoglutathione (GSNO), an endogenous nitric oxide carrier molecule. In cultured rat cortical primary neurons, we observed that GSNO treatment decreased the β-amyloid (Aβ₂₅₋₃₅)-induced pathological tau hyperphosphorylation (Ser396, Ser404, and Ser202/Thr205). The decreased tau hyperphosphorylation correlated with decreased activity of calpain and decreased p35 proteolysis into p25 and Cdk5 activation. GSNO treatment also attenuated the Aβ₂₅₋₃₅-induced activation of GSK-3β which is known to play critical role in tau hyperphosphorylation in addition to Cdk5. Consistent with above studies using cultured neurons, we also observed that systemic GSNO treatment of transgenic mouse model of AD (APPSw/PS1(dE9)) attenuated calpain-mediated p35 proteolysis and Cdk5/GSK-3β activities as well as tau hyperphosphorylation. In addition, GSNO treatment provided neuro- and cognitive protection in APPSw/PS1(dE9) mice. This study describing the GSNO-mediated regulation of tau hyperphosphorylation and cognitive function, for the first time, suggests for therapeutic potential of GSNO as neuro- and cognitive-protective agent for AD.


Brain Research | 2015

S-nitrosoglutathione reduces tau hyper-phosphorylation and provides neuroprotection in rat model of chronic cerebral hypoperfusion.

Je-Seong Won; Balasubramaniam Annamalai; Seungho Choi; Inderjit Singh; Avtar K. Singh

We have previously reported that treatment of rats subjected to permanent bilateral common carotid artery occlusion (pBCCAO), a model of chronic cerebral hypoperfusion (CCH), with S-nitrosoglutathione (GSNO), an endogenous nitric oxide carrier, improved cognitive functions and decreased amyloid-β accumulation in the brains. Since CCH has been implicated in tau hyperphosphorylation induced neurodegeneration, we investigated the role of GSNO in regulation of tau hyperphosphorylation in rat pBCCAO model. The rats subjected to pBCCAO had a significant increase in tau hyperphosphorylation with increased neuronal loss in hippocampal/cortical areas. GSNO treatment attenuated not only the tau hyperphosphorylation, but also the neurodegeneration in pBCCAO rat brains. The pBCCAO rat brains also showed increased activities of GSK-3β and Cdk5 (major tau kinases) and GSNO treatment significantly attenuated their activities. GSNO attenuated the increased calpain activities and calpain-mediated cleavage of p35 leading to production of p25 and aberrant Cdk5 activation. In in vitro studies using purified calpain protein, GSNO treatment inhibited calpain activities while 3-morpholinosydnonimine (a donor of peroxynitrite) treatment increased its activities, suggesting the opposing role of GSNO vs. peroxynitrite in regulation of calpain activities. In pBCCAO rat brains, GSNO treatment attenuated the expression of inducible nitric oxide synthase (iNOS) expression and also reduced the brain levels of nitro-tyrosine formation, thereby indicating the protective role of GSNO in iNOS/nitrosative-stress mediated calpain/tau pathologies under CCH conditions. Taken together with our previous report, these data support the therapeutic potential of GSNO, a biological NO carrier, as a neuro- and cognitive-protective agent under conditions of CCH.


Journal of Neuroscience Research | 2016

Biochemical, cell biological, pathological, and therapeutic aspects of Krabbe's disease.

Je-Seong Won; Avtar K. Singh; Inderjit Singh

Krabbes disease (KD; also called globoid cell leukodystrophy) is a genetic disorder involving demyelination of the central (CNS) and peripheral (PNS) nervous systems. The disease may be subdivided into three types, an infantile form, which is the most common and severe; a juvenile form; and a rare adult form. KD is an autosomal recessive disorder caused by a deficiency of galactocerebrosidase activity in lysosomes, leading to accumulation of galactoceramide and neurotoxic galactosylsphingosine (psychosine [PSY]) in macrophages (globoid cells) as well as neural cells, especially in oligodendrocytes and Schwann cells. This ultimately results in damage to myelin in both CNS and PNS with associated morbidity and mortality. Accumulation of PSY, a lysolipid with detergent‐like properties, over a threshold level could trigger membrane destabilization, leading to cell lysis. Moreover, subthreshold concentrations of PSY trigger cell signaling pathways that induce oxidative stress, mitochondrial dysfunction, apoptosis, inflammation, endothelial/vascular dysfunctions, and neuronal and axonal damage. From the time the “psychosine hypothesis” was proposed, considerable efforts have been made in search of an effective therapy for lowering PSY load with pharmacological, gene, and stem cell approaches to attenuate PSY‐induced neurotoxicity. This Review focuses on the recent advances and prospective research for understanding disease mechanisms and therapeutic approaches for KD.


Redox biology | 2015

S-nitrosoglutathione-mediated STAT3 regulation in efficacy of radiotherapy and cisplatin therapy in head and neck squamous cell carcinoma

Kolanjiappan Kaliyaperumal; Anand K. Sharma; Daniel G. McDonald; Jasdeep S. Dhindsa; Caroline Yount; Avtar K. Singh; Je-Seong Won; Inderjit Singh

S-nitrosoglutathione (GSNO) is an endogenous nitric oxide (NO) carrier that plays a critical role in redox based NO signaling. Recent studies have reported that GSNO regulates the activities of STAT3 and NF-κB via S-nitrosylation dependent mechanisms. Since STAT3 and NF-κB are key transcription factors involved in tumor progression, chemoresistance, and metastasis of head and neck cancer, we investigated the effect of GSNO in cell culture and mouse xenograft models of head and neck squamous cell carcinoma (HNSCC). For the cell culture studies, three HNSCC cell lines were tested (SCC1, SCC14a and SCC22a). All three cell lines had constitutively activated (phosphorylated) STAT3 (Tyr705). GSNO treatment of these cell lines reversibly decreased the STAT3 phosphorylation in a concentration dependent manner. GSNO treatment also decreased the basal and cytokine-stimulated activation of NF-κB in SCC14a cells and reduced the basal low degree of nitrotyrosine by inhibition of inducible NO synthase (iNOS) expression. The reduced STAT3/NF-κB activity by GSNO treatment was correlated with the decreased cell proliferation and increased apoptosis of HNSCC cells. In HNSCC mouse xenograft model, the tumor growth was reduced by systemic treatment with GSNO and was further reduced when the treatment was combined with radiation and cisplatin. Accordingly, GSNO treatment also resulted in decreased levels of phosphorylated STAT3. In summary, these studies demonstrate that GSNO treatment blocks the NF-κB and STAT3 pathways which are responsible for cell survival, proliferation and that GSNO mediated mechanisms complement cispaltin and radiation therapy, and thus could potentiate the therapeutic effect in HNSCC.

Collaboration


Dive into the Avtar K. Singh's collaboration.

Top Co-Authors

Avatar

Inderjit Singh

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Je-Seong Won

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Jinsu Kim

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Seungho Choi

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Balasubramaniam Annamalai

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Nishant Saxena

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Yeong-Bin Im

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Anand K. Sharma

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Anandakumar Shunmugavel

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Mushfiquddin Khan

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge