Network


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

Hotspot


Dive into the research topics where Amareshwar T.K. Singh is active.

Publication


Featured researches published by Amareshwar T.K. Singh.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Biomimetic, synthetic HDL nanostructures for lymphoma

Shuo Yang; Marina G. Damiano; Heng Zhang; Sushant Tripathy; Andrea J. Luthi; Jonathan S. Rink; Andrey Ugolkov; Amareshwar T.K. Singh; Sandeep S. Dave; Leo I. Gordon; C. Shad Thaxton

New therapies that challenge existing paradigms are needed for the treatment of cancer. We report a nanoparticle-enabled therapeutic approach to B-cell lymphoma using synthetic high density lipoprotein nanoparticles (HDL-NPs). HDL-NPs are synthesized using a gold nanoparticle template to control conjugate size and ensure a spherical shape. Like natural HDLs, biomimetic HDL-NPs target scavenger receptor type B-1, a high-affinity HDL receptor expressed by lymphoma cells. Functionally, compared with natural HDL, the gold NP template enables differential manipulation of cellular cholesterol flux in lymphoma cells, promoting cellular cholesterol efflux and limiting cholesterol delivery. This combination of scavenger receptor type B-1 binding and relative cholesterol starvation selectively induces apoptosis. HDL-NP treatment of mice bearing B-cell lymphoma xenografts selectively inhibits B-cell lymphoma growth. As such, HDL-NPs are biofunctional therapeutic agents, whose mechanism of action is enabled by the presence of a synthetic nanotemplate. HDL-NPs are active in B-cell lymphomas and potentially, other malignancies or diseases of pathologic cholesterol accumulation.


Journal of Biological Chemistry | 2009

Blockade of the erbB2 receptor induces cardiomyocyte death through mitochondrial and reactive oxygen species-dependent pathways

Leo I. Gordon; Michael A. Burke; Amareshwar T.K. Singh; Sheila Prachand; Elliot D. Lieberman; Lin Sun; Tejaswitha J Naik; Sathyamangla V. Naga Prasad; Hossein Ardehali

Overexpression of the receptor tyrosine kinase erbB2 (Her2 in humans) is correlated with a poor prognosis in breast and ovarian cancers. Treatment with trastuzumab (a monoclonal antibody against erbB2) improves survival; however, it also causes cardiomyopathy. We hypothesized that blockade of the erbB2 receptor induces cardiomyocyte death through a mitochondrial pathway that is dependent on the production of reactive oxygen species (ROS). We first showed that levels of erbB2 receptor are significantly decreased in an animal model of ischemic heart disease and in human ischemic cardiomyopathy. We treated neonatal rat cardiomyocytes with an inhibitory erbB2 antibody to study the mechanism behind the deleterious effects of erbB2 blockade. These cells displayed a dose-dependent increase in ROS production and cell death compared with control IgG-treated cells; these processes were reversed by the antioxidant, N-acetylcysteine. The effects of erbB2 antibody on both cell death and ROS production were also reversed by cyclosporine A and diazoxide, chemicals that regulate the pro- and anti-apoptotic channels in the mitochondria, respectively. Furthermore, mouse embryonic fibroblasts lacking Bax and Bak (proteins that mediate cell death through a mitochondrial pathway) were resistant to the deleterious effects of erbB2 antibody. These effects of erbB2 blockade appear to occur through a pathway involving AKT and PKC-α. Our results suggest that erbB2 plays a role in cardiomyocyte survival, and that the deleterious effects of trastuzumab on the heart occur through a mitochondrial pathway and is mediated by ROS production. Manipulation of redox signaling may be beneficial in cancer patients receiving trastuzumab.


Nanomedicine: Nanotechnology, Biology and Medicine | 2011

Curcumin nanodisks: formulation and characterization

Mistuni Ghosh; Amareshwar T.K. Singh; Wenwei Xu; Todd Sulchek; Leo I. Gordon; Robert O. Ryan

UNLABELLED Nanodisks (NDs) are nanoscale, disk-shaped phospholipid bilayers whose edge is stabilized by apolipoproteins. In the present study, NDs were formulated with the bioactive polyphenol curcumin at a 6:1 phospholipid-to-curcumin molar ratio. Atomic force microscopy revealed that curcumin-NDs are particles with diameters <50 nm and the thickness of a phospholipid bilayer. When formulated in NDs, curcumin is water soluble and gives rise to a characteristic absorbance spectrum with a peak centered at 420 nm. Fluorescence spectroscopy of curcumin-NDs provided evidence of self-quenching. Incubation of curcumin-NDs with empty NDs relieved the self-quenching, indicating redistribution of curcumin between curcumin-loaded and empty NDs. In HepG2 cells, curcumin-NDs mediated enhanced cell growth inhibition as compared with free curcumin. In a cell culture model of mantle cell lymphoma, curcumin-NDs were a more potent inducer of apoptosis than free curcumin. The nanoscale size of the complexes, combined with their ability to solubilize curcumin, indicates NDs may have in vivo therapeutic applications. FROM THE CLINICAL EDITOR Nanodisks (NDs), disk-shaped phospholipid bilayers stabilized by apolipoproteins, are shown entrap curcumin and improve its delivery to HepG2 and mantle cell lymphoma cells in culture. These novel nanocomplexes demonstrate interesting therapeutic application potentials.


Clinical Cancer Research | 2010

Mitochondrial-mediated apoptosis in lymphoma cells by the diterpenoid lactone andrographolide, the active component of Andrographis paniculata.

Shuo Yang; Andrew M. Evens; Sheila Prachand; Amareshwar T.K. Singh; Savita Bhalla; Kevin David; Leo I. Gordon

Purpose: Andrographolide is a diterpenoid lactone isolated from Andrographis paniculata (King of Bitters), an herbal medicine used in Asia. It has been reported to have anti-inflammatory, antihypertensive, antiviral, and immune-stimulant properties. Furthermore, it has been shown to inhibit cancer cell proliferation and induce apoptosis in leukemia and solid tumor cell lines. Experimental Design: We studied the Burkitt p53-mutated Ramos cell line, the mantle cell lymphoma (MCL) line Granta, the follicular lymphoma (FL) cell line HF-1, and the diffuse large B-cell lymphoma (DLBCL) cell line SUDHL4, as well as primary cells from patients with FL, DLBCL, and MCL. Results: We found that andrographolide resulted in dose- and time-dependent cell death as measured by MTT. Andrographolide significantly increased reactive oxygen species (ROS) production in all cell lines. To determine mechanism of cell death, we measured apoptosis by Annexin V/propidium iodide in the presence and absence of the antioxidant N-acetyl-l-cysteine (NAC), the glutathione (GSH)–depleting agent buthionine sulfoxamine (BSO), or caspase inhibitors. We found that apoptosis was greatly enhanced by BSO, blocked by NAC, and accompanied by poly(ADP-ribose) polymerase cleavage and activation of caspase-3, caspase-8, and caspase-9. We measured BAX conformational change and mitochondrial membrane potential, and using mouse embryonic fibroblast (MEF) Bax/Bak double knockouts (MEFBax−/−/Bak−/−), we found that apoptosis was mediated through mitochondrial pathways, but dependent on caspases in both cell lines and patient samples. Conclusions: Andrographolide caused ROS-dependent apoptosis in lymphoma cell lines and in primary tumor samples, which was enhanced by depletion of GSH and inhibited by NAC or the pan-caspase inhibitor Z-VAD-FMK. Further studies of diterpenoid lactones in lymphoma are warranted. Clin Cancer Res; 16(19); 4755–68. ©2010 AACR.


Cellular Signalling | 2004

Role of protein kinase A, phospholipase C and phospholipase D in parathyroid hormone receptor regulation of protein kinase Cα and interleukin-6 in UMR-106 osteoblastic cells

Julie M. Radeff; Amareshwar T.K. Singh; Paula H. Stern

Parathyroid hormone (PTH) stimulates both bone formation and resorption by activating diverse osteoblast signalling pathways. Upstream signalling for PTH stimulation of protein kinase C-alpha (PKCalpha) membrane translocation and subsequent expression of the pro-resorptive cytokine interleukin-6 (IL-6) was investigated in UMR-106 osteoblastic cells. PTH 1-34, PTH 3-34, PTHrP and PTH 1-31 stimulated PKCalpha translocation and IL-6 promoter activity. Pharmacologic intervention at the adenylyl cyclase (AC) pathway (forskolin, IBMX, PKI) failed to alter PTH 1-34- or PTH 3-34-stimulated PKCalpha translocation. The phosphoinositol-phospholipase C (PI-PLC) antagonist U73122 slightly decreased PTH 1-34-stimulated PKCalpha translocation; however, the control analogue U73343 acted similarly. Propranolol, an inhibitor of phosphatidic acid (PA) phosphohydrolase, decreased diacylglycerol (DAG) formation and attenuated PTH 1-34- and PTH 3-34-stimulated PKCalpha translocation and IL-6 promoter activity, suggesting a phospholipase D (PLD)-dependent mechanism. This is the first demonstration that PLD-mediated signalling leads to both PKC-alpha translocation and IL-6 promoter activation in osteoblastic cells.


British Journal of Haematology | 2008

Hypoxia inducible factor-alpha activation in lymphoma and relationship to the thioredoxin family.

Andrew M. Evens; Paul T. Schumacker; Irene B. Helenowski; Amareshwar T.K. Singh; Danijela Dokic; Anjeni Keswani; Elizabeth Kordeluk; Adekunle Raji; Jane N. Winter; Borko Jovanovic; Arne Holmgren; Beverly P. Nelson; Leo I. Gordon

Hypoxia inducible factors (HIFs) activate oncogenic pathways, while thioredoxins (Trx), including Trx1 and Trx reductases‐1 and ‐2 (TrxR1 and TrxR2), promote HIF‐α stabilization. In immunoblotting studies in lymphoma cell lines we found that Raji and SUDHL4 cells exhibited normoxic HIF‐2α protein stabilization. Five cell lines showed increased TrxR1 expression, while only Namalwa, HF1 and SUDHL4 had Trx1 and TrxR2 activation. Tissue microarrays in diffuse large B‐cell lymphoma (DLBCL) and follicular lymphoma (FL) identified different HIF expression among histological subgroups (e.g. 44% DLBCL vs. 11% of FL cases with moderate‐to‐high expression of HIF‐1α and HIF‐2α, P = 0·0017). These data demonstrate that HIF and the thioredoxin family are abnormally activated in lymphoma.


Endocrinology | 1999

Parathyroid Hormone (PTH)-(1–34),[ Nle8,18,Tyr34]PTH-(3–34) Amide, PTH-(1–31) Amide, and PTH-Related Peptide-(1–34) Stimulate Phosphatidylcholine Hydrolysis in UMR-106 Osteoblastic Cells: Comparison with Effects of Phorbol 12,13-Dibutyrate1

Amareshwar T.K. Singh; Joseph G. Kunnel; Paul J. Strieleman; Paula H. Stern

Studies were performed to determine the effects of PTH and related compounds on phosphatidylcholine (PC) hydrolysis in UMR-106 cells and the pathway by which the PTH effects occurred. The responses were compared with those of phorbol 12,13-dibutyrate (PDBu). Both bovine PTH-(1–34) [bPTH-(1–34)] and PDBu stimulated PC hydrolysis within 10 min. Significant effects were elicited by concentrations of 0.3–1 nm bPTH-(1–34) and 5 nm PDBu. Dose-dependent increases were seen at higher concentrations of both compounds, however, the response to bPTH-(1–34) was reduced at 30 nm. Bovine or human PTH-(1–34) and human PTH-related peptide-(1–34)[ hPTHrP-(1–34)] were equipotent in their effects, whereas bovine[ Nle8,18Tyr34]PTH-(3–34) amide[ bPTH-(3–34)] and hPTH-(1–31) amide [hPTH-(1–31)] were less potent than bPTH-(1–34). bPTH-(3–34) did not antagonize the effects of bPTH-(1–34). Down-regulation of protein kinase C isozymes by 24-h treatment with PDBu completely prevented the stimulatory effect of PDBu on PC hydrolysis,...


British Journal of Haematology | 2010

All trans retinoic acid nanodisks enhance retinoic acid receptor mediated apoptosis and cell cycle arrest in mantle cell lymphoma

Amareshwar T.K. Singh; Andrew M. Evens; Reilly J. Anderson; Jennifer A. Beckstead; Natesan Sankar; Antonella Sassano; Savita Bhalla; Shuo Yang; Leonidas C. Platanias; Trudy M. Forte; Robert O. Ryan; Leo I. Gordon

Mantle cell lymphoma (MCL) is characterized by translocation t(11;14)(q13;q32), aggressive clinical behaviour, and poor patient outcomes following conventional chemotherapy. New treatment approaches are needed that target novel biological pathways. All trans retinoic acid (ATRA) is a key retinoid that acts through nuclear receptors that function as ligand‐inducible transcription factors. The present study evaluated cell killing effects of ATRA‐enriched nanoscale delivery particles, termed nanodisks (ND), on MCL cell lines. Results show that ATRA‐ND induced cell death more effectively than naked ATRA (dimethyl sulphoxide) or empty ND. ATRA‐ND induced reactive oxygen species (ROS) generation to a greater extent than naked ATRA. The antioxidant, N‐acetylcysteine, inhibited ATRA‐ND induced apoptosis. Compared to naked ATRA, ATRA‐ND enhanced G1 growth arrest, up‐regulated p21and p27, and down regulated cyclin D1. At ATRA concentrations that induced apoptosis, expression levels of retinoic acid receptor‐α (RARα) and retinoid X receptor‐γ (RXRγ) were increased. Compared to naked ATRA, ATRA‐ND significantly stimulated transcriptional activity of RARA in a model carcinoma cell line. Furthermore, the RAR antagonist, Ro 41‐5253, inhibited ATRA‐ND induced ROS generation and prevented ATRA‐ND induced cell growth arrest and apoptosis. In summary, incorporation of ATRA into ND enhanced the biological activity of this retinoid in cell culture models of MCL.


Leukemia & Lymphoma | 2011

Curcumin nanodisk-induced apoptosis in mantle cell lymphoma

Amareshwar T.K. Singh; Mistuni Ghosh; Trudy M. Forte; Robert O. Ryan; Leo I. Gordon

Mantle cell lymphoma (MCL) is a pre-germinal center neoplasm characterized by cyclin D1 overexpression resulting from t(11;14)(q13;q32). Since MCL is incurable with standard lymphoma therapies, new treatment approaches are needed that target specific biologic pathways. In the present study, we investigated a novel drug delivery nanovehicle enriched with the bioactive polyphenol, curcumin (curcumin nanodisks; curcumin-ND). Cells treated with curcumin-ND showed a dose-dependent increase in apoptosis. This was accompanied by enhanced generation of reactive oxygen species (ROS). The antioxidant, N-acetylcysteine, inhibited curcumin-ND induced apoptosis, suggesting that ROS generation plays a role in curcumin action on MCL cells. Curcumin-ND decreased cyclin D1, pAkt, pIκBα, and Bcl2 protein. In addition, enhanced FoxO3a and p27 expression as well as caspase-9, -3, and poly(ADP-ribose) polymerase (PARP) cleavage were observed. Curcumin-ND treatment led to enhanced G1 arrest in two cultured cell models of MCL.


Biochimica et Biophysica Acta | 2000

Phosphatidylcholine-specific phospholipase C inhibitor, tricyclodecan-9- yl xanthogenate (D609), increases phospholipase D-mediated phosphatidylcholine hydrolysis in UMR-106 osteoblastic osteosarcoma cells

Amareshwar T.K. Singh; Julie M. Radeff; Joseph G. Kunnel; Paula H. Stern

Our previous studies have shown that parathyroid hormone (PTH) stimulates phosphatidylcholine (PC) hydrolysis by phospholipase D (PLD) and transphosphatidylation in UMR-106 osteoblastic cells. To determine whether phospholipase C (PLC) is also involved in the PTH-mediated PC hydrolysis, we used the inhibitor, tricyclodecan-9-yl xanthogenate (D609), a putatively selective antagonist of this pathway. Consistent with this proposed mechanism, D609 decreased (3)H-phosphocholine in extracts from UMR-106 cells prelabeled with (3)H-choline. Unexpectedly, D609 enhanced PC hydrolysis and transphosphatidylation, suggesting that either there was a compensatory increase in PLD activity when PLC was inhibited, or that D609 directly increased PLD activity. The D609-stimulated increase in PC hydrolysis was rapid, being seen as early as 2 min. The effect of D609 was temperature-sensitive, consistent with an enzymatic mechanism. The D609-stimulated increase in PC hydrolysis was PKC-independent, based upon the lack of effect of down-regulation of PKC by phorbol 12,13-dibutyrate on the response. The studies reveal a novel action of this inhibitor on signaling in osteoblastic cells which might influence downstream responses.

Collaboration


Dive into the Amareshwar T.K. Singh's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shuo Yang

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert O. Ryan

Children's Hospital Oakland Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge