Network


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

Hotspot


Dive into the research topics where Santosh Karnewar is active.

Publication


Featured researches published by Santosh Karnewar.


Diabetes | 2015

Metformin Inhibits Monocyte-to-Macrophage Differentiation via AMPK-Mediated Inhibition of STAT3 Activation: Potential Role in Atherosclerosis

Sathish Babu Vasamsetti; Santosh Karnewar; Anantha Koteswararao Kanugula; Avinash Raj Thatipalli; Jerald Mahesh Kumar; Srigiridhar Kotamraju

Monocyte-to-macrophage differentiation is a critical event that accentuates atherosclerosis by promoting an inflammatory environment within the vessel wall. In this study, we investigated the molecular mechanisms responsible for monocyte-to-macrophage differentiation and, subsequently, the effect of metformin in regressing angiotensin II (Ang-II)-mediated atheromatous plaque formation in ApoE−/− mice. AMPK activity was dose and time dependently downregulated during phorbol myristate acetate (PMA)-induced monocyte-to-macrophage differentiation, which was accompanied by an upregulation of proinflammatory cytokine production. Of note, AMPK activators metformin and AICAR significantly attenuated PMA-induced monocyte-to-macrophage differentiation and proinflammatory cytokine production. However, inhibition of AMPK activity alone by compound C was ineffective in promoting monocyte-to-macrophage differentiation in the absence of PMA. On the other hand, inhibition of c-Jun N-terminal kinase activity inhibited PMA-induced inflammation but not differentiation, suggesting that inflammation and differentiation are independent events. In contrast, inhibition of STAT3 activity inhibited both inflammation and monocyte-to-macrophage differentiation. By decreasing STAT3 phosphorylation, metformin and AICAR through increased AMPK activation caused inhibition of monocyte-to-macrophage differentiation. Metformin attenuated Ang-II–induced atheromatous plaque formation and aortic aneurysm in ApoE−/− mice partly by reducing monocyte infiltration. We conclude that the AMPK-STAT3 axis plays a pivotal role in regulating monocyte-to-macrophage differentiation and that by decreasing STAT3 phosphorylation through increased AMPK activity, AMPK activators inhibit monocyte-to-macrophage differentiation.


PLOS ONE | 2014

Mitochondrial-Targeted Curcuminoids: A Strategy to Enhance Bioavailability and Anticancer Efficacy of Curcumin

Cheruku Apoorva Reddy; Venkateswarlu Somepalli; Trimurtulu Golakoti; Anantha Koteswararao Kanugula; Santosh Karnewar; Karthikraj Rajendiran; Nagarjuna Vasagiri; Sripadi Prabhakar; Periannan Kuppusamy; Srigiridhar Kotamraju; Vijay Kumar Kutala

Although the anti-cancer effects of curcumin has been shown in various cancer cell types, in vitro, pre-clinical and clinical studies showed only a limited efficacy, even at high doses. This is presumably due to low bioavailability in both plasma and tissues, particularly due to poor intracellular accumulation. A variety of methods have been developed to achieve the selective targeting of drugs to cells and mitochondrion. We used a novel approach by conjugation of curcumin to lipophilic triphenylphosphonium (TPP) cation to facilitate delivery of curcumin to mitochondria. TPP is selectively taken up by mitochondria driven by the membrane potential by several hundred folds. In this study, three mitocurcuminoids (mitocurcuminoids-1, 2, and 3) were successfully synthesized by tagging TPP to curcumin at different positions. ESI-MS analysis showed significantly higher uptake of the mitocurcuminoids in mitochondria as compared to curcumin in MCF-7 breast cancer cells. All three mitocurcuminoids exhibited significant cytotoxicity to MCF-7, MDA-MB-231, SKNSH, DU-145, and HeLa cancer cells with minimal effect on normal mammary epithelial cells (MCF-10A). The IC50 was much lower for mitocurcuminoids when compared to curcumin. The mitocurcuminoids induced significant ROS generation, a drop in ΔØm, cell-cycle arrest and apoptosis. They inhibited Akt and STAT3 phosphorylation and increased ERK phosphorylation. Mitocurcuminoids also showed upregulation of pro-apoptotic BNIP3 expression. In conclusion, the results of this study indicated that mitocurcuminoids show substantial promise for further development as a potential agent for the treatment of various cancers.


FEBS Journal | 2014

Statin‐induced inhibition of breast cancer proliferation and invasion involves attenuation of iron transport: intermediacy of nitric oxide and antioxidant defence mechanisms

Anantha Koteswararao Kanugula; Paradesi Naidu Gollavilli; Sathish Babu Vasamsetti; Santosh Karnewar; Raja Gopoju; Ramesh Ummanni; Srigiridhar Kotamraju

Accumulating evidence from in vitro, in vivo, clinical and epidemiological studies shows promising results for the use of statins against many cancers including breast carcinoma. However, the molecular mechanisms responsible for the anti‐proliferative and anti‐invasive properties of statins still remain elusive. In this study, we investigated the involvement of nitric oxide, iron homeostasis and antioxidant defence mechanisms in mediating the anti‐proliferative and anti‐invasive properties of hydrophobic statins in MDA‐MB‐231, MDA‐MB‐453 and BT‐549 metastatic triple negative breast cancer cells. Fluvastatin and simvastatin significantly increased cytotoxicity which was reversed with mevalonate. Interestingly, fluvastatin downregulated transferrin receptor (TfR1), with a concomitant depletion of intracellular iron levels in these cells. Statin‐induced effects were mimicked by geranylgeranyl transferase inhibitor (GGTI‐298) but not farnesyl transferase inhibitor (FTI‐277). Further, it was observed that TfR1 downregulation is mediated by increased nitric oxide levels via inducible nitric oxide synthase (iNOS) expression. NOS inhibitors (asymmetric dimethylarginine and 1400W) counteracted and sepiapterin, a precursor of tetrahydrobiopterin, exacerbated statin‐induced depletion of intracellular iron levels. Notably, fluvastatin increased manganese superoxide dismutase (by repressing the transcription factor DNA damage‐binding protein 2), catalase and glutathione which, in turn, diminished H2O2 levels. Fluvastatin‐induced downregulation of TfR1, matrix metalloproteinase‐2, ‐9 and inhibition of invasion were reversed in the presence of aminotriazole, a specific inhibitor of catalase. Finally, we conclude that fluvastatin, by altering iron homeostasis, nitric oxide generation and antioxidant defence mechanisms, induces triple negative breast cancer cell death.


Nitric Oxide | 2012

Garlic provides protection to mice heart against isoproterenol-induced oxidative damage: Role of nitric oxide

Tarak Nath Khatua; Raju Padiya; Santosh Karnewar; Madhusudana Kuncha; Sachin B. Agawane; Srigiridhar Kotamraju; Sanjay K. Banerjee

Garlic has been widely recognized as a cardioprotective agent. However, the molecular mechanism of its cardioprotective effects is not well established. Here we hypothesized that aqueous garlic homogenate may mediate cardioprotection via nitric oxide (NO). Mice were fed with saline and aqueous garlic homogenate (250 and 500 mgkg(-1)day(-1) orally) for 30 days. In another set of experiment, mice were pre-treated with saline, aqueous garlic homogenate (AGH) (250 mgkg(-1)day(-1) for 30 days), and AGH (30 days) along with L-NAME (20 mgkg(-1)day(-1) i.p. for last 7 days) before inducing acute myocardial infarction by isoproterenol (s.c. injection of isoproterenol 150 mgkg(-1)day(-1) for 2 days) and sacrificed after 48 h. Dose dependent increase in serum NO level was observed after garlic 250 and 500 mgkg(-1) dose feeding. While no change in serum SGPT and SGOT level, a significant decrease in serum LDH level was observed after garlic feeding. Garlic-induced NO formation was further confirmed in human aortic endothelial cells (HAEC). Administration of isoproterenol caused a significant decrease in endogenous antioxidants i.e., myocardial catalase, GSH and GPx activity, and mitochondrial enzyme activities like citrate synthase and β hydroxyacyl CoA dehydrogenase. All those deleterious cardiac changes induced by isoproterenol were significantly attenuated by garlic homogenate. However this beneficial effect of garlic was blunted when garlic was administered with L-NAME, a nonspecific inhibitor of nitric oxide synthase (NOS). Further, a significant increase in myocardial TBARS and decrease in total antioxidant activity was observed in L-NAME treated group compared to isoproterenol treated group. Administration of L-NAME in mice from control group lowered serum and cardiac NO levels without any change of oxidative stress parameters. In conclusion, our study provides novel evidence that garlic homogenate is protective in myocardial infarction via NO-signaling pathway in mice.


Scientific Reports | 2016

Mitochondria-targeted esculetin alleviates mitochondrial dysfunction by AMPK-mediated nitric oxide and SIRT3 regulation in endothelial cells: potential implications in atherosclerosis

Santosh Karnewar; Sathish Babu Vasamsetti; Raja Gopoju; Anantha Koteswararao Kanugula; Sai Krishna Ganji; Sripadi Prabhakar; Nandini Rangaraj; Nitin Tupperwar; Jerald Mahesh Kumar; Srigiridhar Kotamraju

Mitochondria-targeted compounds are emerging as a new class of drugs that can potentially alter the pathophysiology of those diseases where mitochondrial dysfunction plays a critical role. We have synthesized a novel mitochondria-targeted esculetin (Mito-Esc) with an aim to investigate its effect during oxidative stress-induced endothelial cell death and angiotensin (Ang)-II-induced atherosclerosis in ApoE−/− mice. Mito-Esc but not natural esculetin treatment significantly inhibited H2O2- and Ang-II-induced cell death in human aortic endothelial cells by enhancing NO production via AMPK-mediated eNOS phosphorylation. While L-NAME (NOS inhibitor) significantly abrogated Mito-Esc-mediated protective effects, Compound c (inhibitor of AMPK) significantly decreased Mito-Esc-mediated increase in NO production. Notably, Mito-Esc promoted mitochondrial biogenesis by enhancing SIRT3 expression through AMPK activation; and restored H2O2-induced inhibition of mitochondrial respiration. siSIRT3 treatment not only completely reversed Mito-Esc-mediated mitochondrial biogenetic marker expressions but also caused endothelial cell death. Furthermore, Mito-Esc administration to ApoE−/− mice greatly alleviated Ang-II-induced atheromatous plaque formation, monocyte infiltration and serum pro-inflammatory cytokines levels. We conclude that Mito-Esc is preferentially taken up by the mitochondria and preserves endothelial cell survival during oxidative stress by modulating NO generation via AMPK. Also, Mito-Esc-induced SIRT3 plays a pivotal role in mediating mitochondrial biogenesis and perhaps contributes to its anti-atherogenic effects.


Free Radical Biology and Medicine | 2016

Resveratrol attenuates monocyte-to-macrophage differentiation and associated inflammation via modulation of intracellular GSH homeostasis: Relevance in atherosclerosis

Sathish Babu Vasamsetti; Santosh Karnewar; Raja Gopoju; Paradesi Naidu Gollavilli; Sai Ram Narra; Jerald Mahesh Kumar; Srigiridhar Kotamraju

Monocyte-to-macrophage differentiation promotes an inflammatory environment within the arterial vessel wall that causes a mal-adaptive immune response, which contributes to the progression of atheromatous plaque formation. In the current study, we show that resveratrol, a well-known antioxidant, dose-dependently attenuated phorbol myristate acetate (PMA)-induced monocyte-to-macrophage differentiation, as measured by cell adhesion, increase in cell size, and scavenger receptor expression in THP-1 monocytes. Also, resveratrol significantly inhibited PMA-induced pro-inflammatory cytokine/chemokine and matrix metalloprotease (MMP-9) production. This inhibitory effect of resveratrol on monocyte differentiation results from its ability to restore intracellular glutathione (GSH) status, as resveratrol in the presence of buthionine sulfoximine (BSO) failed to affect monocyte differentiation. Furthermore, PMA-induced monocyte differentiation and inflammation was greatly inhibited when cells were co-treated with N-Acetyl-l-cysteine (NAC), a GSH precursor, while the presence of BSO aggravated these processes. These results also show that resveratrol mediated up-regulation of GSH is due to AMP-activated protein kinase (AMPK)-α activation, as compound C (AMPK inhibitor) treatment drastically depleted intracellular GSH and exacerbated PMA-induced monocyte differentiation and pro-inflammatory cytokine production. More importantly, chronic administration of resveratrol efficiently prevented monocyte infiltration and markedly diminished angiotensin (Ang)-II-induced atheromatous plaque formation in apolipoprotein-E knockout (ApoE(-/-)) mice. We conclude that, intracellular GSH status plays a critical role in regulating monocyte-to-macrophage differentiation and inflammation and resveratrol, by restoring GSH levels, inhibits these processes. Taken together, these results suggest that resveratrol can attenuate atherosclerosis, at least, in part, by inhibiting monocyte differentiation and pro-inflammatory cytokines production.


Talanta | 2012

Virtual electrochemical nitric oxide analyzer using copper, zinc superoxide dismutase immobilized on carbon nanotubes in polypyrrole matrix

Thangamuthu Madasamy; Manickam Pandiaraj; Murugesan Balamurugan; Santosh Karnewar; Alby Robson Benjamin; Krishna Arun Venkatesh; Kanagavel Vairamani; Srigiridhar Kotamraju; Chandran Karunakaran

In this work, we have designed and developed a novel and cost effective virtual electrochemical analyzer for the measurement of NO in exhaled breath and from hydrogen peroxide stimulated endothelial cells using home-made potentiostat. Here, data acquisition system (NI MyDAQ) was used to acquire the data from the electrochemical oxidation of NO mediated by copper, zinc superoxide dismutase (Cu,ZnSOD). The electrochemical control programs (graphical user-interface software) were developed using LabVIEW 10.0 to sweep the potential, acquire the current response and process the acquired current signal. The Cu,ZnSOD (SOD1) immobilized on the carbon nanotubes in polypyrrole modified platinum electrode was used as the NO biosensor. The electrochemical behavior of the SOD1 modified electrode exhibited the characteristic quasi-reversible redox peak at the potential, +0.06 V vs. Ag/AgCl. The biological interferences were eliminated by nafion coated SOD1 electrode and then NO was measured selectively. Further, this biosensor showed a wide linear range of response over the concentration of NO from 0.1 μM to 1 mM with a detection limit of 0.1 μM and high sensitivity of 1.1 μA μM(-1). The electroanalytical results obtained here using the developed virtual electrochemical instrument were also compared with the standard cyclic voltammetry instrument and found in agreement with each other.


Biochimica et Biophysica Acta | 2018

Metformin regulates mitochondrial biogenesis and senescence through AMPK mediated H3K79 methylation: Relevance in age-associated vascular dysfunction

Santosh Karnewar; Praveen Kumar Neeli; Devayani Panuganti; Sasikumar Kotagiri; Sreevidya Mallappa; Nishant Jain; Mahesh Kumar Jerald; Srigiridhar Kotamraju

Endothelial senescence in conjunction with mitochondrial dysfunction orchestrates age-associated cardiovascular disorders. In this study we investigated the causal link between these two processes and studied the molecular mechanisms by which metformin acts to coordinate the delay of endothelial senescence via enhancing mitochondrial biogenesis/function. AMPK activators metformin and AICAR delayed endothelial senescence via SIRT1-mediated upregulation of DOT1L, leading to increased trimethylation of H3K79 (H3K79me3). Treatment of cells with either siAMPK or siSIRT1 repressed DOT1L-mediated enhancement of H3K79me3. Moreover, the increase in SIRT3 expression and mitochondrial biogenesis/function by AMPK activators was H3K79me-dependent as H3K79N mutant or siDOT1L abrogated these effects. This was confirmed by the enrichment of H3K79me3 in the SIRT3 promoter with AMPK activation. Intriguingly, enhanced PGC-1α expression by SIRT3 via AMPK activation was responsible for increased hTERT expression and delayed endothelial senescence. In contrast, SIRT3 knockdown caused increased oxidative stress and premature senescence, possibly by depleting hTERT expression. Furthermore, a chronic low dose administration of metformin significantly attenuated vascular aging and inhibited age-associated atherosclerotic plaque formation in ApoE-/- mice. Overall, the results of this study show a novel regulation of mitochondrial biogenesis/function, and cellular senescence by H3K79me acting through SIRT3, thus providing a molecular basis for metformin-mediated age-delaying effects.


FEBS Journal | 2015

AMPK inhibits MTDH expression via GSK3β and SIRT1 activation: potential role in triple negative breast cancer cell proliferation.

Paradesi Naidu Gollavilli; Anantha Koteswararao Kanugula; Rajeswari Koyyada; Santosh Karnewar; Praveen Kumar Neeli; Srigiridhar Kotamraju

Recent studies have highlighted the involvement of metadherin (MTDH), an oncogenic protein, in promoting cancer progression, metastasis and chemoresistance in many cancers including mammary carcinomas. However, the molecular regulation of MTDH is still not completely understood. In this study we document that AMP activated protein kinase (AMPK) activation‐induced anti‐proliferative effects are, in part, mediated by inhibiting MTDH expression in MDA‐MB‐231 and BT‐549 triple negative breast cancer (TNBC) cells. 5‐Aminoimidazole‐4‐carboxamide ribonucleotide (AICAR), an AMPK activator, caused growth arrest, inhibition of migration and invasion of TNBC cells. Intriguingly, AICAR or metformin treatment resulted in significant downregulation of MTDH expression via inhibiting c‐Myc expression. In contrast, treatment of cells with compound C, an inhibitor of AMPK, increased both c‐Myc and MTDH expressions in TNBC cells. Also, AMPK activation caused increased glycogen synthase kinase 3β (GSK3β) activity by inhibiting the inactive phosphorylation at Ser9, on the one hand, and activation of sirtuin1 (SIRT1) by inhibiting Ser47 phosphorylation, as evidenced by deacetylation of p53, on the other hand. Moreover, AMPK‐induced GSK3β and SIRT1 activities were found to be responsible for inhibiting c‐Myc‐mediated upregulation of MTDH, as LiCl (an inhibitor of GSK3β) and EX‐527 (an inhibitor of SIRT1) reversed AICAR‐mediated downregulation of c‐Myc and MTDH expressions. Similar results were observed with siSIRT1 treatment. Furthermore, AICAR and EX‐527 treatments caused increased cell death under MTDH‐depleted conditions. Finally, we uncovered a novel regulation of MTDH expression and showed that AMPK activation by inducing GSK3β and SIRT1 downregulates MTDH expression via inhibiting c‐Myc in TNBC cells.


Journal of Cellular Physiology | 2018

Mitochondria‐targeted esculetin inhibits PAI‐1 levels by modulating STAT3 activation and miR‐19b via SIRT3: Role in acute coronary artery syndrome

Sujana Katta; Santosh Karnewar; Devayani Panuganti; Mahesh Kumar Jerald; B. K. S. Sastry; Srigiridhar Kotamraju

In this study we explored the microRNAs responsible for the regulation of PAI‐1 during LPS‐stimulated inflammation in human aortic endothelial cells and subsequently studied the effect of a newly synthesized mitochondria‐targeted esculetin (Mito‐Esc) that was shown for its anti‐atherosclerotic potential, in modulating PAI‐1 levels and its targeted miRs during angiotensin‐II‐induced atherosclerosis in ApoE−/− mice. LPS‐stimulated PAI‐1 was accompanied with an upregulation of miR‐19b and down‐regulation of miR‐30c. These effects of LPS on PAI‐1 were reversed in the presence of both parent esculetin and Mito‐Esc. However, the effect of Mito‐Esc was more pronounced in the regulation of PAI‐1. In addition, LPS‐stimulated PAI‐1 expression was significantly decreased in cells treated with Anti‐miR‐19b, thereby suggesting that miR‐19b co‐expression plays a key role in PAI‐1 regulation. The results also show that incubation of cells with Stattic, an inhibitor of STAT‐3, inhibited LPS‐stimulated PAI‐1 expression. Interestingly, knockdown of SIRT3, a mitochondrial biogenetic marker, enhanced PAI‐1 levels via modulation of miR‐19b and ‐30c. Mito‐Esc treatment significantly inhibited Ang‐II‐induced PAI‐1, possibly via altering miR‐19b and 30c in ApoE−/− mice. The association between PAI‐1, miR‐19b and ‐30c were further confirmed in plasma and microparticles isolated from patients suffering from acute coronary syndrome of various degrees. Taken together, LPS‐induced PAI‐1 involves co‐expression of miR‐19b and down regulation of miR‐30c, and Mito‐Esc treatment by modulating miR‐19b and miR‐30c through SIRT3 activation, inhibits PAI‐1 levels that, in part, contribute to its anti‐atherosclerotic effects. Moreover, there exists a strong positive correlation between miR‐19b and PAI‐1 in patients suffering from ST‐elevated myocardial infarction.

Collaboration


Dive into the Santosh Karnewar's collaboration.

Top Co-Authors

Avatar

Srigiridhar Kotamraju

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

Anantha Koteswararao Kanugula

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

Sathish Babu Vasamsetti

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

Jerald Mahesh Kumar

Centre for Cellular and Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Paradesi Naidu Gollavilli

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

Raja Gopoju

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

B. Narsaiah

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

C. Kurumurthy

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

Devayani Panuganti

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

Mahesh Kumar Jerald

Centre for Cellular and Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge