Network


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

Hotspot


Dive into the research topics where Jennifer Duplantier is active.

Publication


Featured researches published by Jennifer Duplantier.


Molecular Cancer Therapeutics | 2008

A role for macroautophagy in protection against 4-hydroxytamoxifen-induced cell death and the development of antiestrogen resistance

Julia S. Samaddar; Virgil T. Gaddy; Jennifer Duplantier; Sudharsan Periyasamy Thandavan; Manish Shah; Marlena J. Smith; Jim Rawson; Sylvia B. Smith; John T. Barrett

This study identifies macroautophagy as a key mechanism of cell survival in estrogen receptor–positive (ER+) breast cancer cells undergoing treatment with 4-hydroxytamoxifen (4-OHT). This selective ER modifier is an active metabolite of tamoxifen commonly used for the treatment of breast cancer. Our study provides the following key findings: (a) only 20% to 25% of breast cancer cells treated with 4-OHT in vitro die via caspase-dependent cell death; more typically, the antiestrogen-treated ER+ breast cancer cells express increased levels of macroautophagy and are viable; (b) 4-OHT–induced cell death, but not 4-OHT–induced macroautophagy, can be blocked by the pan-caspase inhibitor z-VAD-fmk, providing strong evidence that these two outcomes of antiestrogen treatment are not linked in an obligatory manner; (c) 4-OHT–resistant cells selected from ER+ breast cancer cells show an increased ability to undergo antiestrogen-induced macroautophagy without induction of caspase-dependent cell death; and (d) 4-OHT, when used in combination with inhibitors of autophagosome function, induces robust, caspase-dependent apoptosis of ER+, 4-OHT–resistant breast cancer cells. To our knowledge, these studies provide the first evidence that macroautophagy plays a critical role in the development of antiestrogen resistance. We propose that targeting autophagosome function will improve the efficacy of hormonal treatment of ER+ breast cancer. [Mol Cancer Ther 2008;7(9):2977–87]


Investigative Ophthalmology & Visual Science | 2008

In vivo protection against retinal neurodegeneration by sigma receptor 1 ligand (+)-pentazocine.

Sylvia B. Smith; Jennifer Duplantier; Ying Dun; Barbara A. Mysona; Penny Roon; Pamela M. Martin; Vadivel Ganapathy

PURPOSE To evaluate the neuroprotective properties of the sigma receptor 1 (sigmaR1) ligand, (+)-pentazocine in an in vivo model of retinal neurodegeneration. METHODS Spontaneously diabetic Ins2(Akita/+) and wild-type mice received intraperitoneal injections of (+)-pentazocine for 22 weeks beginning at diabetes onset. Retinal mRNA and protein were analyzed by RT-PCR and Western blot analysis. Retinal histologic sections were measured to determine total retinal thickness, thicknesses of inner-outer nuclear and plexiform layers (INL, ONL, IPL, INL), and the number of cell bodies in the ganglion cell layer (GCL). Immunolabeling experiments were performed using antibodies specific for 4-hydroxynonenal and nitrotyrosine, markers of lipid peroxidation, and reactive nitrogen species, respectively, and an antibody specific for vimentin to view radial Müller fibers. RESULTS sigmaR1 mRNA and protein levels in the Ins2(Akita/+) retina were comparable to those in the wild-type, indicating that sigmaR1 is an available target during the disease process. Histologic evaluation of eyes of Ins2(Akita/+) mice showed disruption of retinal architecture. By 17 to 25 weeks after birth, Ins2(Akita/+) mice demonstrated approximately 30% and 25% decreases in IPL and INL thicknesses, respectively, and a 30% reduction in ganglion cells. In the (+)-pentazocine-treated group, retinas of Ins2(Akita/+) mice showed remarkable preservation of retinal architecture; IPL and INL thicknesses of (+)-pentazocine-treated Ins2(Akita/+) mouse retinas were within normal limits. The number of ganglion cells was 15.6 +/- 1.5 versus 10.4 +/- 1.2 cells/100 mum retinal length in (+)-pentazocine-treated versus nontreated mutant mice. Levels of nitrotyrosine and 4-hydroxynonenal increased in Ins2(Akita/+) retinas, but were reduced in (+)-pentazocine-treated mice. Retinas of Ins2(Akita/+) mice showed loss of the uniform organization of radial Müller fibers. Retinas of (+)-pentazocine-treated mice maintained the radial organization of glial processes. CONCLUSION Sustained (+)-pentazocine treatment in an in vivo model of retinal degeneration conferred significant neuroprotection, reduced evidence of oxidative stress, and preserved retinal architecture, suggesting that sigmaR1 ligands are promising therapeutic agents for intervention in neurodegenerative diseases of the retina.


Investigative Ophthalmology & Visual Science | 2011

Sigma Receptor 1 Modulates Endoplasmic Reticulum Stress in Retinal Neurons

Yonju Ha; Ying Dun; Muthusamy Thangaraju; Jennifer Duplantier; Zheng Dong; Kebin Liu; Vadivel Ganapathy; Sylvia B. Smith

PURPOSE To investigate the mechanism of σ receptor 1 (σR1) neuroprotection in retinal neurons. METHODS Oxidative stress, which is implicated in diabetic retinopathy, was induced in mouse primary ganglion cells (GCs) and RGC-5 cells, and the effect of the σR1 ligand (+)-pentazocine on pro- and anti-apoptotic and endoplasmic reticulum (ER) stress gene expression was examined. Binding of σR1 to BiP, an ER chaperone protein, and σR1 phosphorylation status were examined by immunoprecipitation. Retinas were harvested from Ins2Akita/+ diabetic mice treated with (+)-pentazocine, and the expression of ER stress genes and of the retinal transcriptome was evaluated. RESULTS Oxidative stress induced the death of primary GCs and RGC-5 cells. The effect was decreased by the application of (+)-pentazocine. Stress increased σR1 binding to BiP and enhanced σR1 phosphorylation in RGC-5 cells. BiP binding was prevented, and σR1 phosphorylation decreased in the presence of (+)-pentazocine. The ER stress proteins PERK, ATF4, ATF6, IRE1α, and CHOP were upregulated in RGC-5 cells during oxidative stress, but decreased in the presence of (+)-pentazocine. A similar phenomenon was observed in retinas of Ins2Akita/+ diabetic mice. Retinal transcriptome analysis of Ins2Akita/+ mice compared with wild-type revealed differential expression of the genes critically involved in oxidative stress, differentiation, and cell death. The expression profile of those genes was reversed when the Ins2Akita/+ mice were treated with (+)-pentazocine. CONCLUSIONS In retinal neurons, the molecular chaperone σR1 binds BiP under stressful conditions; (+)-pentazocine may exert its effects by dissociating σR1 from BiP. As stress in retinal cells increases, phosphorylation of σR1 is increased, which is attenuated when agonists bind to the receptor.


Investigative Ophthalmology & Visual Science | 2009

Endogenous Elevation of Homocysteine Induces Retinal Neuron Death in the Cystathionine-β-Synthase Mutant Mouse

Preethi S. Ganapathy; Brent Moister; Penny Roon; Barbara A. Mysona; Jennifer Duplantier; Ying Dun; Tracy K.V.E. Moister; Marlena J. Farley; Puttur D. Prasad; Kebin Liu; Sylvia B. Smith

PURPOSE To determine the effects of endogenous elevation of homocysteine on the retina using the cystathionine beta-synthase (cbs) mutant mouse. METHODS Retinal homocysteine in cbs mutant mice was measured by high-performance liquid chromatography (HPLC). Retinal cryosections from cbs(-/-) mice and cbs(+/-) mice were examined for histologic changes by light and electron microscopy. Morphometric analysis was performed on retinas of cbs(+/-) mice maintained on a high-methionine diet (cbs(+/-) HM). Changes in retinal gene expression were screened by microarray. RESULTS HPLC analysis revealed an approximate twofold elevation in retinal homocysteine in cbs(+/-) mice and an approximate sevenfold elevation in cbs(-/-) mice. Distinct alterations in the ganglion, inner plexiform, inner nuclear, and epithelial layers were observed in retinas of cbs(-/-) and 1-year-old cbs(+/-) mice. Retinas of cbs(+/-) HM mice demonstrated an approximate 20% decrease in cells of the ganglion cell layer (GCL), which occurred as early as 5-weeks after onset of the HM diet. Microarray analysis revealed alterations in expression of several genes, including increased expression of Aven, Egr1, and Bat3 in retinas of cbs(+/-) HM mice. CONCLUSIONS This study provides the first analysis of morphologic and molecular effects of endogenous elevations of retinal homocysteine in an in vivo model. Increased retinal homocysteine alters inner and outer retinal layers in cbs homozygous mice and older cbs heterozygous mice, and it primarily affects the cells of the GCL in younger heterozygous mice. Elevated retinal homocysteine alters expression of genes involved in endoplasmic reticular stress, N-methyl-d-aspartate (NMDA) receptor activation, cell cycle, and apoptosis.


Journal of Neurochemistry | 2008

Serine racemase expression and d-serine content are developmentally regulated in neuronal ganglion cells of the retina

Ying Dun; Jennifer Duplantier; Penny Roon; Pamela M. Martin; Vadivel Ganapathy; Sylvia B. Smith

d‐Serine, the endogenous ligand for the glycine modulatory binding site of the NMDA receptor, and serine racemase, the enzyme that converts l‐serine to d‐serine, have been reported in vertebrate retina; initial reports suggested that localization was restricted to Müller glial cells. Recent reports, in which d‐serine and serine racemase were detected in neurons of the brain, prompted the present investigation of neuronal expression of d‐serine and serine racemase in retina and whether expression patterns were developmentally regulated. RT‐PCR, in situ hybridization, western blotting, immunohistochemistry, and immunocytochemical methods were used to localize d‐serine and serine racemase in intact retina obtained from 1 to 3 day, 3 week, and 18 week mouse retinas and in primary ganglion cells harvested by immunopanning from neonatal mouse retina. Results of these analyses revealed robust expression of d‐serine and serine racemase in ganglion cells, both in intact retina and in cultured cells. The levels appear to be developmentally regulated with d‐serine levels being quite high in ganglion cells of neonatal retinas and decreasing rapidly postnatally. Serine racemase levels are also developmentally regulated, with high levels detected during the early postnatal period, but diminishing considerably in the mature retina. This represents the first report of neuronal expression of d‐serine and serine racemase in the vertebrate retina and suggests an important contribution of neuronal d‐serine during retinal development.


Investigative Ophthalmology & Visual Science | 2010

Molecular and biochemical characterization of folate transport proteins in retinal müller cells

B. Renee Bozard; Preethi S. Ganapathy; Jennifer Duplantier; Barbara A. Mysona; Yonju Ha; Penny Roon; Robert G. Smith; I. David Goldman; Puttur D. Prasad; Pamela M. Martin; Vadivel Ganapathy; Sylvia B. Smith

PURPOSE To analyze the mechanisms of folate uptake in retinal Müller cells. METHODS RT-PCR and Western blot analysis were performed in freshly isolated neural retina and RPE/eyecup, primary mouse Müller cells, and rMC-1 cells for the three known folate transport proteins folate receptor alpha (FRalpha), proton-coupled folate transporter (PCFT), and reduced folate carrier (RFC). Laser scanning confocal microscopy (LSCM) and immunoelectron microscopy were used to determine the subcellular location of FRalpha and PCFT in primary Müller cells. The pH dependence of the uptake of [(3)H]-methyltetrahydrofolate ([(3)H]-MTF) was assayed in Müller cells in the presence/absence of thiamine pyrophosphate, an inhibitor of RFC. RESULTS FRalpha and PCFT are expressed abundantly in the retina in several cell layers, including the inner nuclear layer; they are present in primary mouse Müller cells and rMC-1 cells. LSCM localized these proteins to the plasma membrane, nuclear membrane, and perinuclear region. Immunoelectron microscopic studies revealed the colocalization of FRalpha and PCFT on the plasma membrane and nuclear membrane and within endosomal structures. Müller cell uptake of [(3)H]-MTF was robust at pH 5.0 to 6.0, consistent with PCFT activity, but also at neutral pH, reflecting RFC function. RFC was expressed in mouse Müller cells that had been allowed to proliferate in culture, but not in freshly isolated primary cells. CONCLUSIONS FRalpha and PCFT are expressed in retinal Müller cells and colocalize in the endosomal compartment, suggesting that the two proteins may work coordinately to mediate folate uptake. The unexpected finding of RFC expression and activity in cultured Müller cells may reflect the upregulation of this protein under proliferative conditions.


Current Eye Research | 2010

Sensitivity of staurosporine-induced differentiated RGC-5 cells to homocysteine

Preethi S. Ganapathy; Ying Dun; Yonju Ha; Jennifer Duplantier; John Bradley Allen; Amina Farooq; B. Renee Bozard; Sylvia B. Smith

Purpose: Homocysteine is implicated in ganglion cell death associated with glaucoma. To understand mechanisms of homocysteine-induced cell death, we analyzed the sensitivity of the RGC-5 cell line, differentiated using staurosporine, to physiologically-relevant levels of the excitotoxic amino acid homocysteine. Methods: RGC-5 cells were differentiated 24 hr using 316 nM staurosporine and tested for expression of Thy 1.2 via immunodetection, RT-PCR, and immunoblotting. The sensitivity of staurosporine-differentiated RGC-5 cells to physiological levels of homocysteine (50, 100, 250 μM) and to high levels of homocysteine (1 mM), glutamate (1 mM), and oxidative stress (25 μM:10 mU/ml xanthine:xanthine oxidase) was assessed by TUNEL assay and by immunodetection of cleaved caspase-3. The sensitivity of undifferentiated RGC-5 cells to high (1, 5, and 10 mM) homocysteine was also examined. Results: Undifferentiated RGC-5 cells express Thy 1.2 mRNA and protein. Staurosporine-differentiated RGC-5 cells extend neurite processes and express Thy 1.2 after 24 hr differentiation; they express NF-L after 1 and 3 days differentiation. Treatment of staurosporine -differentiated RGC-5 cells with 50, 100, or 250 µM homocysteine did not alter neurite processes nor induce cell death (detected by TUNEL and active caspase-3) to a level greater than that observed in the control (non-homocysteine-treated, staurosporine-differentiated) cells. The 1 mM dosage of homocysteine in staurosporine-differentiated RGC-5 cells also did not induce cell death above control levels, although 18 hr treatment of non-differentiated RGC-5 cells with 5 mM homocysteine decreased survival by 50%. Conclusions: RGC-5 cells differentiated for 24 hr with 316 nM staurosporine project robust neurite processes and are positive for ganglion cell markers consistent with a more neuronal phenotype than non-staurosporine-differentiated RGC-5 cells. However, concentrations of homocysteine known to induce ganglion cell death in vivo and in primary ganglion cells are not sufficient to induce death of RGC-5 cells, even when they are differentiated with staurosporine.


Investigative Ophthalmology & Visual Science | 2008

Expression and Function of System N Glutamine Transporters (SN1/SN2 or SNAT3/SNAT5) in Retinal Ganglion Cells

Nagavedi S. Umapathy; Ying Dun; Pamela M. Martin; Jennifer Duplantier; Penny Roon; Puttur D. Prasad; Sylvia B. Smith; Vadivel Ganapathy

PURPOSE Glutamine transport is essential for the glutamate-glutamine cycle, which occurs between neurons and glia. System N, consisting of SN1 (SNAT3) and SN2 (SNAT5), is the principal mediator of glutamine transport in retinal Müller cells. Mediators of glutamine transport in retinal ganglion cells were investigated. METHODS The relative contributions of various transport systems for glutamine uptake (systems N, A, L, y+L, ASCT, and ATB(0,+)) were examined in RGC-5 cells based on differential features of the individual transport systems. mRNA for the genes encoding members of these transport systems were analyzed by RT-PCR. Based on these data, SN1 and SN2 were analyzed in mouse retina, RGC-5 cells, and primary mouse ganglion cells (GCs) by in situ hybridization (ISH), immunofluorescence (IF), and Western blotting. RESULTS Three transport systems--N, A, and L--participated in glutamine uptake in RGC-5 cells. System N was the principal contributor; systems A and L contributed considerably less. ISH and IF revealed SN1 and SN2 expression in the ganglion, inner nuclear, and photoreceptor cell layers. SN1 and SN2 colocalized with the ganglion cell marker Thy 1.2 and with the Müller cell marker vimentin, confirming their presence in both retinal cell types. SN1 and SN2 proteins were detected in primary mouse GCs. CONCLUSIONS These findings suggest that in addition to its role in glutamine uptake in retinal glial cells, system N contributes significantly to glutamine uptake in ganglion cells and, hence, contributes to the retinal glutamate-glutamine cycle.


Cell and Tissue Research | 2009

Effects of hyperglycemia and oxidative stress on the glutamate transporters GLAST and system xc- in mouse retinal Müller glial cells.

Barbara A. Mysona; Ying Dun; Jennifer Duplantier; Vadivel Ganapathy; Sylvia B. Smith


Investigative Ophthalmology & Visual Science | 2010

Sigma receptor 1 modulates ER stress in retinal neurons

Yonju Ha; Ying Dun; M. Thangaraju; Jennifer Duplantier; Dong Zheng; Kela Liu; Vadivel Ganapathy; Stephen R. Smith

Collaboration


Dive into the Jennifer Duplantier's collaboration.

Top Co-Authors

Avatar

Sylvia B. Smith

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Ying Dun

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Vadivel Ganapathy

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Barbara A. Mysona

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Penny Roon

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Pamela M. Martin

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Puttur D. Prasad

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Yonju Ha

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

B. Renee Bozard

Georgia Regents University

View shared research outputs
Researchain Logo
Decentralizing Knowledge