Network


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

Hotspot


Dive into the research topics where Rajalakshmi Veeranan-Karmegam is active.

Publication


Featured researches published by Rajalakshmi Veeranan-Karmegam.


Nature Communications | 2015

DNMT1 is essential for mammary and cancer stem cell maintenance and tumorigenesis

Rajneesh Pathania; Selvakumar Elangovan; Ravi Padia; Pengyi Yang; Senthilkumar Cinghu; Rajalakshmi Veeranan-Karmegam; Pachiappan Arjunan; Jaya P. Gnana-Prakasam; Fulzele Sadanand; Lirong Pei; Chang Sheng Chang; Jeong Hyeon Choi; Huidong Shi; Santhakumar Manicassamy; Puttur D. Prasad; Suash Sharma; Vadivel Ganapathy; Raja Jothi; Muthusamy Thangaraju

Mammary stem/progenitor cells (MaSCs) maintain self-renewal of the mammary epithelium during puberty and pregnancy. DNA methylation provides a potential epigenetic mechanism for maintaining cellular memory during self-renewal. Although DNA methyltransferases (DNMTs) are dispensable for embryonic stem cell maintenance, their role in maintaining MaSCs and cancer stem cells (CSCs) in constantly replenishing mammary epithelium is unclear. Here we show that DNMT1 is indispensable for MaSC maintenance. Furthermore, we find that DNMT1 expression is elevated in mammary tumors, and mammary gland-specific DNMT1 deletion protects mice from mammary tumorigenesis by limiting the CSC pool. Through genome-scale methylation studies, we identify ISL1 as a direct DNMT1 target, hypermethylated and downregulated in mammary tumors and CSCs. DNMT inhibition or ISL1 expression in breast cancer cells limits CSC population. Altogether, our studies uncover an essential role for DNMT1 in MaSC and CSC maintenance and identify DNMT1-ISL1 axis as a potential therapeutic target for breast cancer treatment.


Investigative Ophthalmology & Visual Science | 2012

GPR109A as an anti-inflammatory receptor in retinal pigment epithelial cells and its relevance to diabetic retinopathy.

Deeksha Gambhir; Sudha Ananth; Rajalakshmi Veeranan-Karmegam; Selvakumar Elangovan; Shanterian Hester; Eric Jennings; Stefan Offermanns; Julian Nussbaum; Sylvia B. Smith; Muthusamy Thangaraju; Vadivel Ganapathy; Pamela M. Martin

PURPOSE Retinal pigment epithelium (RPE) expresses GPR109A, a receptor for the vitamin niacin and the ketone body β-hydroxybutyrate (β-HB). Because diabetes results in elevated levels of β-HB, here we studied expression of the receptor in diabetic retina. We also investigated its functional relevance in RPE. METHODS Retinal expression of GPR109A in diabetic mice and postmortem human eyes was evaluated by quantitative PCR (qPCR). ARPE-19 cells and primary wild-type and Gpr109a(-/-) mouse RPE cells were exposed to TNF-α in the presence or absence of niacin or β-HB, followed by analysis of IL-6 and Ccl2 expression via real-time qPCR and ELISA. RESULTS GPR109A expression was increased in diabetic mouse and human retina. TNF-α increased the expression and secretion of IL-6 and Ccl2 in ARPE-19 cells. Niacin and β-HB suppressed these effects, implicating GPR109A as the target responsible for mediation of the observed effects. Primary RPE cells from wild-type mice behaved similarly. In contrast, GPR109A ligands failed to suppress TNF-α-induced expression and secretion of IL-6 and Ccl2 in primary RPE cells from Gpr109a(-/-) mice, confirming that the observed anti-inflammatory effects were mediated specifically by Gpr109a. CONCLUSIONS GPR109A plays an anti-inflammatory role in RPE and its expression is upregulated in diabetes. Inflammation is a key causative factor in the pathogenesis of diabetic retinopathy. We speculate that the increased expression of GPR109A and elevation of its ligand β-HB in diabetes are mechanisms by which the tissue attempts to fight inflammation in this disease. Pharmacological activation of GPR109A may therefore have therapeutic potential in clinical management of diabetic retinopathy.


Journal of Neurochemistry | 2008

Cystamine prevents haloperidol-induced decrease of BDNF/TrkB signaling in mouse frontal cortex

Anilkumar Pillai; Rajalakshmi Veeranan-Karmegam; Krishnan M. Dhandapani; Sahebarao P. Mahadik

The role of brain‐derived neurotrophic factor (BDNF) has been implicated in the pathophysiology as well as treatment outcome of schizophrenia. Rodent studies indicate that several antipsychotic drugs have time‐dependent (and differential) effects on BDNF levels in the brain. Earlier studies from our laboratory have indicated that long‐term treatment with haloperidol (HAL) decreases BDNF, reduced GSH and anti‐apoptotic marker, Bcl‐xl protein levels and increases the expression of pro‐apoptotic proteins in rat frontal cortex. Furthermore, findings from human as well as rodent studies suggest that treatment of schizophrenia must involve the neuroprotective strategies to improve the neuropathology and thereby clinical outcome. In the present study, we investigated the potential of cystamine (CYS), an anti‐oxidant and anti‐apoptotic compound, to prevent HAL‐induced reduction in BDNF, GSH, and Bcl‐xl protein levels in mice and the signaling mechanism(s) involved in the beneficial effects of CYS. The results indicated that CYS as well as cysteamine (the FDA‐approved precursor of CYS) increased BDNF protein levels in mouse frontal cortex 7 days after treatment. CYS co‐treatment prevented chronic HAL treatment‐induced reduction in BDNF, GSH, and Bcl‐xl protein levels. CYS treatment enhanced TrkB‐tyrosine phosphorylation and activated Akt and extracellular signal‐regulated kinase (ERK)1/2, downstream molecules of TrkB signaling. In addition, in vitro experiments with mouse cortical neurons showed that CYS prevented the HAL‐induced reduction in neuronal cell viability and BDNF protein levels, and increase in apoptosis. BDNF‐neutralizing antibody as well as K252a, a selective inhibitor of neurotrophin signaling blocked the CYS‐mediated neuroprotection. Moreover, CYS‐mediated neuroprotection is also blocked by LY294002, a phosphatidylinositol 3‐kinase inhibitor or PD98059, a mitogen‐activated protein kinase kinase (MEK) inhibitor. Thus, CYS protects cortical neurons through a mechanism involving TrkB receptor activation, and a signaling pathway involving phosphatidylinositol 3‐kinase and MAPK. The findings from the present study may be helpful for the development of novel neuroprotective strategies to improve the treatment outcome of schizophrenia.


Investigative Ophthalmology & Visual Science | 2011

Polarized Distribution of Heme Transporters in Retinal Pigment Epithelium and Their Regulation in the Iron-Overload Disease Hemochromatosis

Jaya P. Gnana-Prakasam; Sushma K. Reddy; Rajalakshmi Veeranan-Karmegam; Sylvia B. Smith; Pamela M. Martin; Vadivel Ganapathy

PURPOSE FLVCR, BCRP, and PCFT/HCP-1 represent the three heme transporters identified thus far in mammalian cells, but there is very little known about their expression and regulation in the retina. In this study, the expression of these transporters in mouse retina and retinal pigment epithelium (RPE) and their regulation in the iron-overload disease hemochromatosis were examined. METHODS The expression of FLVCR, BCRP, and PCFT in mouse retina and primary mouse RPE cells was studied by RT-PCR and immunofluorescence. Polarized localization of the transporters in RPE was studied by co-localization using a specific marker of the RPE apical membrane. Uptake of heme in primary RPE cells was determined using zinc-mesoporphyrin, a fluorescent heme analogue. The regulation of heme transporters by iron overload was studied in two genetic models of hemochromatosis (HFE-null mouse and HJV-null mouse) and in two nongenetic models of iron overload (cytomegalovirus infection and treatment with ferric ammonium citrate). RESULTS All three heme transporters were expressed in the retina and RPE. In the RPE, the expression of FLVCR was restricted to the apical membrane, and the expression of BCRP and PCFT was restricted to the basolateral membrane. In all cases of iron overload, the expression of FLVCR and PCFT was upregulated and that of BCRP was downregulated. CONCLUSIONS Hemochromatosis is associated not only with excessive accumulation of free iron in the retina and RPE but also with excessive accumulation of heme. Since heme is toxic at high levels, as is free iron, heme-induced oxidative damage may also play a role in hemochromatosis-associated retinal pathology.


Biochimica et Biophysica Acta | 2014

Regulation of the cholesterol efflux transporters ABCA1 and ABCG1 in retina in hemochromatosis and by the endogenous siderophore 2,5-dihydroxybenzoic acid.

Sudha Ananth; Jaya P. Gnana-Prakasam; Yangzom D. Bhutia; Rajalakshmi Veeranan-Karmegam; Pamela M. Martin; Sylvia B. Smith; Vadivel Ganapathy

Hypercholesterolemia and polymorphisms in the cholesterol exporter ABCA1 are linked to age-related macular degeneration (AMD). Excessive iron in retina also has a link to AMD pathogenesis. Whether these findings mean a biological/molecular connection between iron and cholesterol is not known. Here we examined the relationship between retinal iron and cholesterol using a mouse model (Hfe(-/-)) of hemochromatosis, a genetic disorder of iron overload. We compared the expression of the cholesterol efflux transporters ABCA1 and ABCG1 and cholesterol content in wild type and Hfe(-/-) mouse retinas. We also investigated the expression of Bdh2, the rate-limiting enzyme in the synthesis of the endogenous siderophore 2,5-dihydroxybenzoic acid (2,5-DHBA) in wild type and Hfe(-/-) mouse retinas, and the influence of this siderophore on ABCA1/ABCG1 expression in retinal pigment epithelium. We found that ABCA1 and ABCG1 were expressed in all retinal cell types, and that their expression was decreased in Hfe(-/-) retina. This was accompanied with an increase in retinal cholesterol content. Bdh2 was also expressed in all retinal cell types, and its expression was decreased in hemochromatosis. In ARPE-19 cells, 2,5-DHBA increased ABCA1/ABCG1 expression and decreased cholesterol content. This was not due to depletion of free iron because 2,5-DHBA (a siderophore) and deferiprone (an iron chelator) had opposite effects on transferrin receptor expression and ferritin levels. We conclude that iron is a regulator of cholesterol homeostasis in retina and that removal of cholesterol from retinal cells is impaired in hemochromatosis. Since excessive cholesterol is pro-inflammatory, hemochromatosis might promote retinal inflammation via cholesterol in AMD.


Journal of Cell Science | 2016

A new isoform of Drosophila non-muscle Tropomyosin 1 interacts with Kinesin-1 and functions in oskar mRNA localization

Rajalakshmi Veeranan-Karmegam; Devi Prasad Boggupalli; Guojun Liu; Graydon B. Gonsalvez

ABSTRACT Recent studies have revealed that diverse cell types use mRNA localization as a means to establish polarity. Despite the prevalence of this phenomenon, much less is known regarding the mechanism by which mRNAs are localized. The Drosophila melanogaster oocyte provides a useful model for examining the process of mRNA localization. oskar (osk) mRNA is localized at the posterior of the oocyte, thus restricting the expression of Oskar protein to this site. The localization of osk mRNA is microtubule dependent and requires the plus-end-directed motor Kinesin-1. Unlike most Kinesin-1 cargoes, localization of osk mRNA requires the Kinesin heavy chain (Khc) motor subunit, but not the Kinesin light chain (Klc) adaptor. In this report, we demonstrate that a newly discovered isoform of Tropomyosin 1, referred to as Tm1C, directly interacts with Khc and functions in concert with this microtubule motor to localize osk mRNA. Apart from osk mRNA localization, several additional Khc-dependent processes in the oocyte are unaffected upon loss of Tm1C. Our results therefore suggest that the Tm1C–Khc interaction is specific for the osk localization pathway. Highlighted Article: A previously uncharacterized isoform of Tropomyosin, referred to as Tm1C, directly interacts with Kinesin heavy chain and functions along with this microtubule motor to localize oskar mRNA in Drosophila oocytes.


Journal of Immunology | 2015

Amino Acid Metabolism Inhibits Antibody-Driven Kidney Injury by Inducing Autophagy

Kapil Chaudhary; Rahul Shinde; Haiyun Liu; Jaya P. Gnana-Prakasam; Rajalakshmi Veeranan-Karmegam; Lei Huang; Buvana Ravishankar; Jillian Bradley; Nino Kvirkvelia; Malgorzata McMenamin; Wei Xiao; Daniel T. Kleven; Andrew L. Mellor; Michael P. Madaio; Tracy L. McGaha

Inflammatory kidney disease is a major clinical problem that can result in end-stage renal failure. In this article, we show that Ab-mediated inflammatory kidney injury and renal disease in a mouse nephrotoxic serum nephritis model was inhibited by amino acid metabolism and a protective autophagic response. The metabolic signal was driven by IFN-γ–mediated induction of indoleamine 2,3-dioxygenase 1 (IDO1) enzyme activity with subsequent activation of a stress response dependent on the eIF2α kinase general control nonderepressible 2 (GCN2). Activation of GCN2 suppressed proinflammatory cytokine production in glomeruli and reduced macrophage recruitment to the kidney during the incipient stage of Ab-induced glomerular inflammation. Further, inhibition of autophagy or genetic ablation of Ido1 or Gcn2 converted Ab-induced, self-limiting nephritis to fatal end-stage renal disease. Conversely, increasing kidney IDO1 activity or treating mice with a GCN2 agonist induced autophagy and protected mice from nephritic kidney damage. Finally, kidney tissue from patients with Ab-driven nephropathy showed increased IDO1 abundance and stress gene expression. Thus, these findings support the hypothesis that the IDO–GCN2 pathway in glomerular stromal cells is a critical negative feedback mechanism that limits inflammatory renal pathologic changes by inducing autophagy.


Investigative Ophthalmology & Visual Science | 2013

Induction of the Cystine/Glutamate Exchanger SLC7A11 in Retinal Pigment Epithelial Cells by the Antipsoriatic Drug Monomethylfumarate

Sudha Ananth; Ellappan Babu; Rajalakshmi Veeranan-Karmegam; Brooke R. Bozard Baldowski; Thomas Boettger; Pamela M. Martin

PURPOSE Oxidative stress is a common pathological factor in degenerative retinal diseases; therefore, identifying novel strategies for its limitation is critically important and highly relevant clinically. Along these lines, our present goal was to evaluate the effect(s) of the fumarate ester and antipsoriatic agent monomethylfumarate (MMF) on the expression and functional activity of the cystine/glutamate exchanger SLC7A11 (system xc(-)), a transport system critical to potentiation of antioxidant signaling in retina. METHODS ARPE-19 and primary mouse RPE cells were cultured in the presence or absence of varying concentrations of MMF (0-5000 μM) for 0 to 24 hours. MMF (10 mM) was also delivered intravitreally to mouse eyes. RT-PCR, radiolabeled uptake, Western blotting, and glutathione (GSH) assays were then used to evaluate the effects of MMF on endogenous antioxidant machinery. RESULTS MMF induced system xc(-), Nrf2, and hypoxia-inducible factor 1α (Hif-1α) in cultured RPE cells. Additionally, the compound was recognized as a transportable substrate by the Na(+)-coupled monocarboxylate transporter SLC5A8 (SMCT1). In vivo these factors were evidenced by a significant increase in retinal levels of GSH. CONCLUSIONS MMF stimulates multiple pathways in retinal cells that potentiate cellular events leading to the upregulation of genes/mechanisms that function to protect retina against various forms of insult; upregulation of system xc(-) is one such consequence. To our knowledge, this is the first report that fumarate esters, compounds already employed clinically for other indications, are effective in retina via xc(-) induction. This novel, hitherto unknown mechanism helps to explain the antioxidant feature of these compounds and highlights their therapeutic potential in retina.


Investigative Ophthalmology & Visual Science | 2011

Transport via Slc5a8 (SMCT1) is Obligatory for 2-Oxothiazolidine-4-carboxylate to Enhance Glutathione Production in Retinal Pigment Epithelial Cells

Ellappan Babu; Sudha Ananth; Rajalakshmi Veeranan-Karmegam; Veena Coothankandaswamy; Sylvia B. Smith; Thomas Boettger; Vadivel Ganapathy; Pamela M. Martin

PURPOSE To evaluate the role of SLC5A8 in the transport of 2-oxothiazolidine-4-carboxylate (OTC) and to determine whether OTC augments glutathione production in RPE cells, thereby providing protection against oxidative stress. METHODS SLC5A8-mediated transport of OTC was monitored in Xenopus laevis oocytes by electrophysiological means. Saturation kinetics, Na(+)-activation kinetics, and inhibition by ibuprofen were analyzed by monitoring OTC-induced currents as a measure of transport activity. Oxidative stress was induced in ARPE-19 cells and primary RPE cells isolated from wild type and Slc5a8(-/-) mouse retinas using H(2)O(2), and the effects of OTC on cell death and intracellular glutathione concentration were examined. RESULTS Heterologous expression of human SLC5A8 in X. laevis oocytes induced Na(+)-dependent inward currents in the presence of OTC under voltage-clamp conditions. The transport of OTC via SLC5A8 was saturable, with a K(t) of 104 ± 3 μM. The Na(+)-activation kinetics was sigmoidal with a Hill coefficient of 1.9 ± 0.1, suggesting involvement of two Na(+) in the activation process. Ibuprofen, a blocker of SLC5A8, inhibited SLC5A8-mediated OTC transport; the concentration necessary for half-maximal inhibition was 17 ± 1 μM. OTC increased glutathione levels and protected ARPE-19 and primary RPE cells isolated from wild type mouse retinas from H(2)O(2)-induced cell death. These effects were abolished in primary RPE isolated from Slc5a8(-/-) mouse retinas. CONCLUSIONS OTC is a transportable substrate for SLC5A8. OTC augments glutathione production in RPE cells, thereby protecting them from oxidative damage. Transport via SLC5A8 is obligatory for this process.


Investigative Ophthalmology & Visual Science | 2013

Loss of Hfe leads to progression of tumor phenotype in primary retinal pigment epithelial cells.

Jaya P. Gnana-Prakasam; Rajalakshmi Veeranan-Karmegam; Veena Coothankandaswamy; Sushma K. Reddy; Pamela M. Martin; Muthusamy Thangaraju; Sylvia B. Smith; Vadivel Ganapathy

PURPOSE Hemochromatosis is a disorder of iron overload arising mostly from mutations in HFE. HFE is expressed in retinal pigment epithelium (RPE), and Hfe(-/-) mice develop age-related iron accumulation and retinal degeneration associated with RPE hyperproliferation. Here, the mechanism underlying the hyperproliferative phenotype in RPE was investigated. METHODS Cellular senescence was monitored by β-galactosidase activity. Gene expression was monitored by real-time PCR. Survivin was analyzed by Western blot and immunofluorescence. Migration and invasion were monitored using appropriate kits. Glucose transporters (GLUTs) were monitored by 3-O-methyl-D-glucose uptake. Histone deacetylases (HDACs) were studied by monitoring catalytic activity and acetylation status of histones H3/H4. RESULTS Hfe(-/-) RPE cells exhibited slower senescence rate and higher survivin expression than wild type cells. Hfe(-/-) cells migrated faster and showed greater glucose uptake and increased expression of GLUTs. The expression of HDACs and DNA methyltransferase (DNMTs) also was increased. Similarly, RPE cells from hemojuvelin (Hjv)-knockout mice, another model of hemochromatosis, also had increased expression of GLUTs, HDACs, and DNMTs. The expression of Slc5a8 was decreased in Hfe(-/-) RPE cells, but treatment with a DNA methylation inhibitor restored the transporter expression, indicating involvement of DNA methylation in the silencing of Slc5a8 in Hfe(-/-) cells. CONCLUSIONS RPE cells from iron-overloaded mice exhibit several features of tumor cells: decreased senescence, enhanced migration, increased glucose uptake, and elevated levels of HDACs and DNMTs. These features are seen in Hfe(-/-) RPE cells as well as in Hjv(-/-) RPE cells, providing a molecular basis for the hyperproliferative phenotype of Hfe(-/-) and Hjv(-/-) RPE cells.

Collaboration


Dive into the Rajalakshmi Veeranan-Karmegam's collaboration.

Top Co-Authors

Avatar

Vadivel Ganapathy

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Pamela M. Martin

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Sudha Ananth

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Sylvia B. Smith

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wanwisa Promsote

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Deeksha Gambhir

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amany Tawfik

Georgia Regents University

View shared research outputs
Researchain Logo
Decentralizing Knowledge