Network


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

Hotspot


Dive into the research topics where Preethi S. Ganapathy is active.

Publication


Featured researches published by Preethi S. Ganapathy.


Investigative Ophthalmology & Visual Science | 2011

The Role of N-Methyl-D-Aspartate Receptor Activation in Homocysteine-Induced Death of Retinal Ganglion Cells

Preethi S. Ganapathy; Richard E. White; Yonju Ha; B. Renee Bozard; Paul L. McNeil; R. William Caldwell; Sanjiv Kumar; Stephen M. Black; Sylvia B. Smith

PURPOSE Elevated plasma homocysteine has been implicated in glaucoma, a vision disorder characterized by retinal ganglion cell death. The toxic potential of homocysteine to ganglion cells is known, but the mechanisms are not clear. A mechanism of homocysteine-induced death of cerebral neurons is via N-methyl-D-aspartate (NMDA) receptor overstimulation, leading to excess calcium influx and oxidative stress. This study examined the role of the NMDA receptor in homocysteine-mediated ganglion cell death. METHODS Primary mouse ganglion cells were used for these experiments. NMDA receptor stimulation by homocysteine was determined by patch clamp analysis and fluorescent detection of intracellular calcium. NMDA receptor involvement in homocysteine-mediated cell death was determined through assessment of lactate dehydrogenase release and TUNEL analysis. These experiments used the NMDA receptor blocker MK-801. Induction of reactive species superoxide, nitric oxide, and peroxynitrite was measured by electron paramagnetic resonance spectroscopy, chemiluminescent nitric oxide detection, and immunoblotting for nitrotyrosine, respectively. RESULTS 50 μM homocysteine stimulated the NMDA receptor in presence of 100 μM glycine. Homocysteine induced 59.67 ± 4.89% ganglion cell death that was reduced to 19.87 ± 3.03% with cotreatment of 250 nM MK-801. Homocysteine elevated intracellular calcium ∼7-fold, which was completely prevented by MK-801. Homocysteine treatment increased superoxide and nitric oxide levels by ∼40% and ∼90%, respectively, after 6 hours. Homocysteine treatment elevated peroxynitrite by ∼85% after 9 hours. CONCLUSIONS These experiments provide compelling evidence that homocysteine induces retinal ganglion cell toxicity through direct NMDA receptor stimulation and implicate, for the first time, the induction of oxidative stress as a potent mechanism of homocysteine-mediated ganglion cell death.


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.


Experimental Eye Research | 2012

Age-related changes in visual function in cystathionine-beta-synthase mutant mice, a model of hyperhomocysteinemia

Minzhong Yu; Gwen M. Sturgill-Short; Preethi S. Ganapathy; Amany Tawfik; Neal S. Peachey; Sylvia B. Smith

Homocysteine is an amino acid required for the metabolism of methionine. Excess homocysteine is implicated in cardiovascular and neurological disease and new data suggest a role in various retinopathies. Mice lacking cystathionine-beta-synthase (cbs(-/-)) have an excess of retinal homocysteine and develop anatomical abnormalities in multiple retinal layers, including photoreceptors and ganglion cells; heterozygous (cbs(+/-)) mice demonstrate ganglion cell loss and mitochondrial abnormalities in the optic nerve. The purpose of the present study was to determine whether elevated homocysteine, due to absent or diminished cbs, alters visual function. We examined cbs(-/-) (3 weeks) and cbs(+/-) mice (5, 10, 15, 30 weeks) and results were compared to those obtained from wild type (WT) littermates. Conventional dark- and light-adapted ERGs were recorded, along with dc-ERG to assess retinal pigment epithelial (RPE) function. The visual evoked potential (VEP) was used to assess transmission to the visual cortex. The amplitudes of the major ERG components were reduced in cbs(-/-) mice at age 3 weeks and VEPs were delayed markedly. These findings are consistent with the early retinal disruption observed anatomically in these mice. In comparison, at 3 weeks of age, responses of cbs(+/-) mice did not differ significantly from those of WT mice. Functional abnormalities were not observed in cbs(+/-) mice until 15 weeks of age, at which time amplitude reductions were noted for the ERG a- and b-wave and the light peak component, but not for other components generated by the RPE. VEP implicit times were delayed in cbs(+/-) mice at 15 and 30 weeks, while VEP amplitudes were unaffected. The later onset of functional defects in cbs(+/-) mice is consistent with a slow loss of ganglion cells reported previously in the heterozygous mutant. Light peak abnormalities indicate that RPE function is also compromised in older cbs(+/-) mice. The data suggest that severe elevations of homocysteine are associated with marked alterations of retinal function while modest homocysteine elevation is reflected in milder and delayed alterations of retinal function. The work lays the foundation to explore the role of homocysteine in retinal diseases such as glaucoma and optic neuropathy.


Investigative Ophthalmology & Visual Science | 2011

Homocysteine-Mediated Modulation of Mitochondrial Dynamics in Retinal Ganglion Cells

Preethi S. Ganapathy; Richard L. Perry; Amany Tawfik; Robert M. Smith; Elizabeth Perry; Penny Roon; B. Renee Bozard; Yonju Ha; Sylvia B. Smith

PURPOSE To evaluate the effect of excess homocysteine on the regulation of retinal ganglion cell mitochondrial dynamics. METHODS Mice deficient in cystathionine-β-synthase (cbs) were used as a model of hyperhomocysteinemia. Gene and protein expression analyses of Opa1 and Fis1 were performed on cbs⁺/⁻ neural retinas. Mitochondria within retinal ganglion cell axons underwent systematic ultrastructural analysis to measure area, length, width, and the distance between the mitochondria and the axon wall. Primary mouse ganglion cells were cultured, treated with homocysteine, and assessed for levels of Opa1 and Fis1 protein, the number of mitochondria per length of neurite, and levels of cleaved caspase-3. RESULTS Opa1 and Fis1 protein levels in cbs⁺/⁻ neural retinas were elevated to 191.00% ± 26.40% and 226.20% ± 4.57%, respectively, compared with wild-type. Mitochondria of cbs⁺/⁻ retinas were smaller in all parameters studied, including area (0.32 ± 0.01 μm² vs. 0.42 ± 0.02 μm²), compared with wild-type. Primary ganglion cells treated with homocysteine had elevations in Opa1 and Fis1 proteins, a significantly higher number of mitochondria per length of neurite (0.1781 ± 0.017 vs. 0.1156 ± 0.012), and significantly higher levels of cleaved caspase-3 compared with control. CONCLUSIONS This study provides the first evidence that homocysteine-induced ganglion cell loss involves the dysregulation of mitochondrial dynamics, both in vivo and in vitro. The present data suggest increased mitochondrial fission as a novel mechanism of homocysteine toxicity to neurons. Of particular relevance are glaucoma and Alzheimers disease, neurodegenerative diseases that are associated with hyperhomocysteinemia and, more recently, have implicated increased mitochondrial fission in their pathogeneses.


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 | 2013

Alterations of Retinal Vasculature in Cystathionine-Beta-Synthase Mutant Mice, a Model of Hyperhomocysteinemia

Amany Tawfik; Mohamed Al-Shabrawey; Penny Roon; Srinivas Sonne; Jason Covar; Surapoon Matragoon; Preethi S. Ganapathy; Sally S. Atherton; Azza B. El-Remessy; Vadivel Ganapathy; Sylvia B. Smith

PURPOSE Mice with moderate/severe hyperhomocysteinemia due to deficiency or absence of the cbs gene encoding cystathionine-beta-synthase (CBS) have marked retinal disruption, ganglion cell loss, optic nerve mitochondrial dysfunction, and ERG defects; those with mild hyperhomocysteinemia have delayed retinal morphological/functional phenotype. Excess homocysteine is a risk factor for cardiovascular diseases; however, it is not known whether excess homocysteine alters retinal vasculature. METHODS Cbs(+/+), cbs(+/-), and cbs(-/-) mice (age ∼3 weeks) were subjected to angiography; retinas were harvested for cryosections, flat-mount preparations, or trypsin digestion and subjected to immunofluorescence microscopy to visualize vessels using isolectin-B4, to detect angiogenesis using anti-VEGF and anti-endoglin (anti-CD105) and activated glial cells (anti-glial fibrillary acidic protein [anti-GFAP]) and to investigate the blood-retinal barrier using the tight junction markers zonula occludens-1 (ZO-1) and occludin. Expression of vegf was determined by quantitative RT-PCR (qRT-PCR) and immunoblotting. Human retinal endothelial cells (HRECs) were treated with excess homocysteine to analyze permeability. RESULTS Angiography revealed vascular leakage in cbs(-/-) mice; immunohistochemical analysis demonstrated vascular patterns consistent with ischemia; isolectin-B4 labeling revealed a capillary-free zone centrally and new vessels with capillary tufts midperipherally. This was associated with increased vegf mRNA and protein, CD105, and GFAP in cbs(-/-) retinas concomitant with a marked decrease in ZO-1 and occludin. Homocysteine-treated HRECs showed increased permeability. CONCLUSIONS Severe elevation of homocysteine in cbs(-/-) mutant mice is accompanied by alterations in retinal vasculature (ischemia, neovascularization, and incompetent blood-retinal barrier). The marked disruption of retinal structure and decreased visual function reported in cbs(-/-) mice may reflect vasculopathy as well as neuropathy.


Ophthalmology and Eye Diseases | 2009

Diabetes Accelerates Retinal Neuronal Cell Death In A Mouse Model of Endogenous Hyperhomocysteinemia.

Preethi S. Ganapathy; Penny Roon; Tracy K.V.E. Moister; Barbara A. Mysona; Sylvia B. Smith

Hyperhomocysteinemia has been implicated in visual dysfunction. We reported recently that mice with endogenous hyperhomocysteinemia, due to mutation of the cystathionine-β-synthase (cbs) gene, demonstrate loss of neurons in the retinal ganglion cell (RGC) layer and other retinal layers as homocysteine levels increase. Some clinical studies implicate hyperhomocysteinemia in the pathogenesis of diabetic retinopathy, which is also characterized by RGC loss. The present study used cbs+/– mice to determine whether modest elevation of plasma homocysteine, in the presence of diabetes, accelerates neuronal cell loss. Diabetes (DB) was induced in 3 wk old cbs+/– and wildtype mice using streptozotocin; four groups of mice were studied: DB cbs+/– non-DB cbs+/– DB cbs+/+; non-DB cbs+/+. One group of diabetic cbs+/– mice was maintained on a high methionine diet (HMD, 0.5% methionine drinking water) to increase plasma homocysteine slightly. Eyes were harvested at 5, 10 and 15 weeks post-onset of diabetes; retinal cryosections were examined by light microscopy and subjected to systematic morphometric analysis. Diabetic cbs+/– had significantly fewer RGCs at 5 weeks compared to age-matched, non-diabetic cbs+/– and wildtype controls (10.0 ± 0.5 versus 14.9 ± 0.5 and 15.8 ± 0.6 cells/100 μm retina length, respectively). Significant differences in retinas of DB/high homocysteine versus controls were obtained 15 wks post-onset of diabetes including fewer RGCS and decreased thickness of inner nuclear and plexiform layers. Moderate increases in plasma homocysteine coupled with diabetes cause a more dramatic alteration of retinal phenotype than elevated homocysteine or diabetes alone and suggest that diabetes accelerates the retinal neuronal death in hyperhomocysteinemic mice.


Journal of Pharmaceutical Sciences | 2012

Transport of the Synthetic Opioid Peptide DADLE ([d-Ala2,d-Leu5]–Enkephalin) in Neuronal Cells

Sudha Ananth; Santoshanand V. Thakkar; Jaya P. Gnana-Prakasam; Pamela M. Martin; Preethi S. Ganapathy; Sylvia B. Smith; Vadivel Ganapathy

The sodium-coupled oligopeptide transporters 1 and 2 (SOPT1 and SOPT2) transport peptides consisting of at least five amino acids and show potential for the delivery of therapeutically relevant peptides/peptidomimetics. Here, we examined the expression of these two transporters in the retinal neuronal cell line RGC-5. These cells showed robust uptake activity for the synthetic pentapeptide DADLE ([D-Ala(2),D-Leu(5)]-Enkephalin). The uptake was Na(+) dependent and saturable (K(t), 6.2 ± 0.6 μM). A variety of oligopeptides inhibited DADLE uptake. The uptake of the competing oligopeptides was directly demonstrated with fluorescein isothiocyanate-labeled Tyr-Gly-Gly-Phe-Leu-Arg-Arg-Ile-Arg-Pro-Lys-Leu-Lys in RGC-5 cells and primary mouse retinal ganglion cells. The characteristics of DADLE uptake matched those of SOPT2. We then examined the expression of SOPT1 in these cells with deltorphin II (Tyr-D-Ala-Phe-Glu-Val-Val-Gly-NH(2)) as the substrate and found that RGC-5 cells also expressed SOPT1. As it is already known that SOPT1 is expressed in the neuronal cell line SK-N-SH, we investigated SOPT2 expression in these cells to determine whether the presence of both oligopeptide transporters is a common feature of neuronal cells. These studies showed that SK-N-SH cells also expressed SOPT2. This constitutes the first report on the functional characterization of SOPT1 and SOPT2 in retinal neuronal cells and on the expression of SOPT2 in nonretinal neuronal cells.


Handbook of Nutrition, Diet and the Eye | 2014

Folate Transport in Retina and Consequences on Retinal Structure and Function of Hyperhomocysteinemia

Sylvia B. Smith; Preethi S. Ganapathy; R.B. Bozard; V. Ganapathy

Folate is a water-soluble vitamin that is essential for the synthesis of DNA, RNA, and some amino acids. It is required for the proper function of every cell, including those of the retina. This chapter summarizes reported studies of the transport mechanisms by which retinal cells take up folate, including folate receptor α, reduced folate carrier, and proton-coupled folate transporter. It is well known that when folate is deficient, homocysteine levels increase. Genetic mutations can also trigger hyperhomocysteinemia. The second portion of the chapter focuses on the consequences on retina structure and function under conditions of hyperhomocysteinemia. It describes studies in several models in which defects in enzymes associated with the remethylation and transsulfuration pathway are associated with varying degrees of retinal neuropathy and vasculopathy.

Collaboration


Dive into the Preethi S. Ganapathy's collaboration.

Top Co-Authors

Avatar

Sylvia B. Smith

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Penny Roon

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

Vadivel Ganapathy

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

B. Renee Bozard

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Barbara A. Mysona

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

V. Ganapathy

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Amany Tawfik

Georgia Regents University

View shared research outputs
Researchain Logo
Decentralizing Knowledge