Network


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

Hotspot


Dive into the research topics where Savita Bhalla is active.

Publication


Featured researches published by Savita Bhalla.


Clinical Cancer Research | 2009

PCI-24781 Induces Caspase and Reactive Oxygen Species–Dependent Apoptosis Through NF-κB Mechanisms and Is Synergistic with Bortezomib in Lymphoma Cells

Savita Bhalla; Sriram Balasubramanian; Kevin David; Mint Sirisawad; Joseph J. Buggy; Lauren Mauro; Sheila Prachand; Richard J. Miller; Leo I. Gordon; Andrew M. Evens

Purpose: We investigated the cytotoxicity and mechanisms of cell death of the broad-spectrum histone deacetylase (HDAC) inhibitor PCI-24781, alone and combined with bortezomib in Hodgkin lymphoma and non-Hodgkin lymphoma cell lines and primary lymphoproliferative (CLL/SLL) cells. Experimental Design: Apoptosis, mitochondrial membrane potential, cell cycle analysis, and reactive oxygen species (ROS) were measured by flow cytometry, whereas caspase activation was determined by Western blot. Nuclear factor κB (NF-κB)-related mRNAs were quantified by reverse transcription-PCR, NF-κB–related proteins by Western blotting, and NF-κB DNA-binding activity by electromobility shift assay. Finally, gene expression profiling was analyzed. Results: PCI-24781 induced concentration-dependent apoptosis that was associated with prominent G0/G1 arrest, decreased S-phase, increased p21 protein, and increased ROS in Hodgkin lymphoma and non-Hodgkin lymphoma cell lines. Dose-dependent apoptosis with PCI-24781 was also seen among primary CLL/SLL cells. PCI-24781–induced apoptosis was shown to be ROS- and caspase-dependent. Combined PCI-24781/bortezomib treatment resulted in strong synergistic apoptosis in all non-Hodgkin lymphoma lines (combination indices, 0.19-0.6) and was additive in Hodgkin lymphoma and primary CLL/SLL cells. Further, PCI-24781/bortezomib resulted in increased caspase cleavage, mitochondrial depolarization, and histone acetylation compared with either agent alone. Gene expression profiling showed that PCI-24781 alone significantly down-regulated several antioxidant genes, proteasome components, and NF-κB pathway genes, effects that were enhanced further with bortezomib. Reverse transcription-PCR confirmed down-regulation of NF-κB1 (p105), c-Myc, and IκB-kinase subunits, where NF-κB DNA binding activity was decreased. Conclusion: We show that PCI-24781 results in increased ROS and NF-κB inhibition, leading to caspase-dependent apoptosis. We also show that bortezomib is synergistic with PCI-24781. This combination or PCI-24781 alone has potential therapeutic value in lymphoma.


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.


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.


Blood | 2011

The novel anti-MEK small molecule AZD6244 induces BIM-dependent and AKT-independent apoptosis in diffuse large B-cell lymphoma

Savita Bhalla; Andrew M. Evens; Bojie Dai; Sheila Prachand; Leo I. Gordon; Ronald B. Gartenhaus

The RAS/RAF/MEK/ERK signaling pathway has been largely unexplored as a potential therapeutic target in lymphoma. The novel 2nd generation anti-MEK small molecule, AZD6244, down-regulated its direct downstream target, phospho-ERK (pERK) in germinal center and nongerminal center diffuse large B-cell lymphoma (DLBCL) cell lines and primary cells. Similar decreased pERK levels were noted despite constitutive activation (CA) of MEK. Consequently, several lymphoma-related ERK substrates were down-regulated by AZD6244 including MCT-1, c-Myc, Bcl-2, Mcl-1, and CDK1/2. AZD6244 induced time- and dose-dependent antiproliferation and apoptosis in all DLBCL cell lines and fresh/primary cells (IC(50) 100nM-300nM). Furthermore, AZD6244 resulted in significantly less tumor compared with control in an in vivo DLBCL SCID xenograft model. Cell death was associated with cleaved PARP, caspases-8, -9, and -3, and apoptosis was caspase-dependent. In addition, there was stabilization of FoxO3a, activation of BIM and PUMA, and a significant decrease in c-Myc transcripts. Moreover, siRNA knockdown of BIM abrogated AZD6244-related apoptosis, while shRNA knockdown of ERK minimally sensitized cells. Finally, manipulation of AKT with transfection of OCI-LY3 cells with CA-AKT or through chemical inhibition (LY294002) had minimal effect on AZD6244-induced cell death. Altogether, these findings show that the novel anti-MEK agent, AZD6244, induced apoptosis in DLBCL and that cell death was BIM-dependent.


British Journal of Haematology | 2010

Glutathione depletion enhances arsenic trioxide-induced apoptosis in lymphoma cells through mitochondrial-independent mechanisms

Savita Bhalla; Leo I. Gordon; Kevin David; Sheila Prachand; Amareshwar T.K. Singh; Shuo Yang; Jane N. Winter; Dongsheng Guo; Thomas V. O'Halloran; Leonidas C. Platanias; Andrew M. Evens

Arsenic trioxide (ATO) is an effective therapeutic agent for acute promyelocytic leukemia (APL) (Evens et al, 2004). In APL, ATO induces differentiation at low concentrations, while inducing apoptosis at higher concentrations (Miller, et al 2002). In addition, ATO-induced apoptosis in APL is mediated through the mitochondrial apoptotic pathway, resulting in part from the production of reactive oxygen species (ROS) such as hydrogen peroxide (Dai, et al 1999, Yi, et al 2002). High intracellular levels of glutathione (GSH) confer resistance to ATO in part through the detoxification of ROS. Compounds that promote ROS and/or deplete protective metabolites such as GSH are able to sensitize tumor cells to oxidative cytolysis. Buthionine sulfoximine (BSO), a selective inhibitor of gamma glutamylcysteine synthetase, is known to effectively deplete cellular GSH (Davison, et al 2003, Gartenhaus, et al 2002). We evaluated herein the cytotoxic activity and cell death pathways induced by ATO alone and combined with BSO in non-Hodgkin’s lymphoma (NHL) cell lines and primary lymphoproliferative cells.


Clinical Cancer Research | 2014

The Novel Organic Arsenical Darinaparsin Induces MAPK-Mediated and SHP1-Dependent Cell Death in T-cell Lymphoma and Hodgkin Lymphoma Cells and Human Xenograft Models

Dashnamoorthy Ravi; Savita Bhalla; Ronald B. Gartenhaus; Jennifer Crombie; Irawati Kandela; Jaya Sharma; Andrew P. Mazar; Andrew M. Evens

Purpose: Darinaparsin (Zio-101) is a novel organic arsenical compound with encouraging clinical activity in relapsed/refractory T-cell lymphoma (TCL) and Hodgkin lymphoma (HL); however, little is known about its mechanism of action. Experimental Design: TCL cell lines (Jurkat, Hut78, and HH) and HL cell lines (L428, L540, and L1236) were examined for in vitro cell death by MTT assay and Annexin V–based flow cytometry. Jurkat and L540-derived xenografts in SCID mice were examined for in vivo tumor inhibition and survival. Biologic effects of darinaparsin on the MAPK pathway were investigated using pharmacologic inhibitors, RNAi and transient transfection for overexpression for SHP1 and MEK. Results: Darinaparsin treatment resulted in time- and dose-dependent cytotoxicity and apoptosis in all TCL and HL cell lines. In addition, darinaparsin had more rapid, higher, and sustained intracellular arsenic levels compared with arsenic trioxide via mass spectrometry. In vivo experiments with Jurkat (TCL) and L540 (HL)-derived lymphoma xenografts showed significant inhibition of tumor growth and improved survival in darinaparsin-treated SCID mice. Biologically, darinaparsin caused phosphorylation of ERK (and relevant downstream substrates) primarily by decreasing the inhibitory SHP1 phosphatase and coimmunoprecipitation showed significant ERK/SHP1 interaction. Furthermore, ERK shRNA knockdown or constitutive overexpression of SHP1 resulted in increased apoptosis, whereas cotreatment with pharmacologic MEK inhibitors resulted in synergistic cell death. Conversely, SHP1 blockade (via pharmacologic inhibition or RNAi) and MEK constitutive activation decreased darinaparsin-related cell death. Conclusions: Altogether, these data show that darinaparsin is highly active in HL and TCL and its activity is dependent primarily on MAPK mechanisms. Clin Cancer Res; 20(23); 6023–33. ©2014 AACR.


PLOS ONE | 2013

Paradoxical regulation of hypoxia inducible factor-1α (HIF-1α) by histone deacetylase inhibitor in diffuse large B-cell lymphoma.

Savita Bhalla; Andrew M. Evens; Sheila Prachand; Paul T. Schumacker; Leo I. Gordon

Hypoxia inducible factor (HIF) is important in cancer, as it regulates various oncogenic genes as well as genes involved in cell survival, proliferation, and migration. Elevated HIF-1 protein promotes a more aggressive tumor phenotype, and greater HIF-1 expression has been demonstrated to correlate with poorer prognosis, increased risk of metastasis and increased mortality. Recent reports suggest that HIF-1 activates autophagy, a lysosomal degradation pathway which may promote tumor cell survival. We show here that HIF-1α expression is constitutively active in multiple diffuse large B cell lymphoma (DLBCL) cell lines under normoxia and it is regulated by the PI3K/AKT pathway. PCI-24781, a pan histone deacetylase inhibitor (HDACI), enhanced accumulation of HIF-1α and induced autophagy initially, while extended incubation with the drug resulted in inhibition of HIF-1α. We tested the hypothesis that PCI-24781- induced autophagy is mediated by HIF-1α and that inhibition of HIF-1α in these cells results in attenuation of autophagy and decreased survival. We also provide evidence that autophagy serves as a survival pathway in DLBCL cells treated with PCI-24781 which suggests that the use of autophagy inhibitors such as chloroquine or 3-methyl adenine in combination with PCI-24781 may enhance apoptosis in lymphoma cells.


Cancer Biology & Therapy | 2016

Functional characterization of NAD dependent de-acetylases SIRT1 and SIRT2 in B-Cell Chronic Lymphocytic Leukemia (CLL)

Savita Bhalla; Leo I. Gordon

ABSTRACT Sirtuins (SIRT) are nicotinamide adenine dinucleotide (NAD+) dependent deacetylases or ADP- ribosyl transferases (ARTs) that deacetylate lysine residues on various proteins regulating a variety of cellular and metabolic processes. These enzymes regulate metabolism, cell survival, differentiation and DNA repair. SIRT proteins play an important role in the survival and drug resistance of cancer cells. The purpose of the present study was to investigate the expression and role of SIRT in chronic lymphocytic leukemia (CLL). We analyzed the expression of SIRT1 and SIRT2 in CLL and normal B cells using the Oncomine database as well as by Western blotting of fresh CLL cells from patients and pro-lymphocytic leukemia (PLL) cell lines, JVM-3 and MEC-2. We showed that both primary CLL cells and JVM-3 and MEC-2 cell lines overexpress high levels of functional SIRT1 and SIRT2. SIRT inhibitors EX-527 and sirtinol impair cell growth, induce ROS production, loss of mitochondrial membrane potential and apoptosis in primary CLL cells and cell lines. Using shRNA knock down of SIRT1 and SIRT2 in JVM-3 and MEC-2 cell lines, we showed that expression of both proteins is crucial for the survival of these cells. Furthermore, studies in nutrient deprived conditions suggest a role of SIRT in metabolism in CLL. These results demonstrate that the inhibition of SIRT1 and SIRT2 activity may be a new therapeutic approach for CLL.


Molecular Cancer Therapeutics | 2013

Abstract B201: Targeting HIF-1α in diffuse large B cell lymphoma: Role for HDAC inhibitors.

Savita Bhalla; Andrew M. Evens; Sheila Prachand; Paul T. Schumacker; Leo I. Gordon

Hypoxia inducible factor (HIF) is important in cancer, as it regulates various oncogenic genes as well as genes involved in cell survival, proliferation, and migration. Elevated HIF-1 protein promotes a more aggressive tumor phenotype, and greater HIF-1 expression has been demonstrated to correlate with poorer prognosis, increased risk of metastasis and increased mortality. Recent reports suggest that HIF-1 activates autophagy, a lysosomal degradation pathway which may promote tumor cell survival. We show here that HIF-1α is constitutively active in multiple diffuse large B cell lymphoma (DLBCL) cell lines under normoxia and it is regulated by the PI3K/AKT pathway. PCI-24781, a pan histone deacetylase inhibitor (HDACI), enhanced accumulation of HIF-1α and induced autophagy initially, while extended incubation with the drug resulted in inhibition of HIF-1α. We tested the hypothesis that PCI-24781- induced autophagy is mediated by HIF-1α and that inhibition of HIF-1α in these cells results in attenuation of autophagy and decreased survival. We also provide evidence that autophagy serves as a survival pathway in DLBCL cells treated with PCI-24781 which suggest that the use of autophagy inhibitors such as chloroquine or 3-methyl adenine in combination with PCI-24781 may enhance apoptosis in lymphoma cells. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):B201. Citation Format: Savita Bhalla, Andrew M. Evens, Sheila Prachand, Paul T. Schumacker, Leo I. Gordon. Targeting HIF-1α in diffuse large B cell lymphoma: Role for HDAC inhibitors. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr B201.


Cancer Research | 2012

Abstract 4735: Epigenetic modification of the PI3K/Akt/HIF1α pathway in lymphoma cells by HDAC inhibition

Savita Bhalla; Andrew M. Evens; Shuo Yang; Sheila Prachand; Leo I. Gordon

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Hypoxia-inducible factors (HIFs) strongly influence many cellular process such as erythropoiesis, angiogenesis, energy metabolism, pH regulation, cell migration and tumor invasion. Under normoxic conditions, HIF-1α possesses a very short half-life and is rapidly degraded by the ubiquitin proteasome pathway and remains inactive. In hypoxia, HIF-1α is stabilized and forms a complex with HIF-1α that allows HIF-1 to function as a transcription factor. HIF-1α is activated only during hypoxia under normal physiologic conditions. By contrast, HIF-1α is frequently activated in cancer cells, including under normoxic conditions by oncogene products or impaired activity of tumor suppressor genes. Recent studies have shown that the expression of HIF1α is increased in a variety of human tumors a significant fraction of diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma patients, implicating a potential role of dysregulated HIF activation. Constitutive expression of HIF-1α enhances vascularization, increases glucose metabolism, and induces the expression of anti-apoptotic proteins in cancer tissues, thus making it an attractive target for cancer therapy. We hypothesized that epigenetic modification of lymphoma cells by histone deacetylase (HDAC) inhibition may modify HIF-1α expression and lead to cell death. We therefore studied the role of the pan HDACi PCI-24781 (PCI) in HIF1α mediated cell death. Western blot analysis of cells treated with 0.125μM to 0.5μM of PCI show significant down regulation of HIF1α protein in all NHL cell lines. To confirm that down regulation of HIF1α is an important factor in cell apoptosis induced by PCI, we incubated cells with the small molecule inhibitor of HIF1α, PX-478 in combination with PCI. Combination of PCI and PX-478 at concentrations of 0.5μM and 10μM respectively, showed an increase in apoptosis as compared to either agent alone in all the DLBCL cell lines. Moreover, knocking down HIF1α in SUDHL4 and OCI-LY3 cell lines using shRNA followed by treatment with PCI also showed the similar effect. We further show that down regulation of HIF1α by PCI is mediated via the PI3K/Akt pathway and inhibition of the PI3K/Akt pathway by LY294002 inhibits HIF1α protein expression. Furthermore pre-incubation of cells with LY inhibits PCI-induced apoptosis. By contrast, constitutive activation of AKT using Myr-Akt-CA shows an increase in HIF1α protein and partial attenuation of PCI-induced apoptosis.We conclude that the activation of the PI3K/Akt/HIF-1α pathway plays a critical role in mediating hypoxia-induced drug resistance leading to unfavorable treatment outcome in lymphoma. Our study suggests a mechanism by which lymphoma cells become resistant to HDAC inhibitors. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4735. doi:1538-7445.AM2012-4735

Collaboration


Dive into the Savita Bhalla'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

Kevin David

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge