Network


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

Hotspot


Dive into the research topics where Debashree Basudhar is active.

Publication


Featured researches published by Debashree Basudhar.


Biochimica et Biophysica Acta | 2009

The Emergence of Nitroxyl (HNO) as a Pharmacological Agent

Christopher H. Switzer; Wilmarie Flores-Santana; Danellie Mancardi; Sonia Donzelli; Debashree Basudhar; Lisa A. Ridnour; Katrina M. Miranda; Jon M. Fukuto; Nazerano Paolocci; David A. Wink

Once a virtually unknown nitrogen oxide, nitroxyl (HNO) has emerged as a potential pharmacological agent. Recent advances in the understanding of the chemistry of HNO has led to the an understanding of HNO biochemistry which is vastly different from the known chemistry and biochemistry of nitric oxide (NO), the one-electron oxidation product of HNO. The cardiovascular roles of NO have been extensively studied, as NO is a key modulator of vascular tone and is involved in a number of vascular related pathologies. HNO displays unique cardiovascular properties and has been shown to have positive lusitropic and ionotropic effects in failing hearts without a chronotropic effect. Additionally, HNO causes a release of CGRP and modulates calcium channels such as ryanodine receptors. HNO has shown beneficial effects in ischemia reperfusion injury, as HNO treatment before ischemia-reperfusion reduces infarct size. In addition to the cardiovascular effects observed, HNO has shown initial promise in the realm of cancer therapy. HNO has been demonstrated to inhibit GAPDH, a key glycolytic enzyme. Due to the Warburg effect, inhibiting glycolysis is an attractive target for inhibiting tumor proliferation. Indeed, HNO has recently been shown to inhibit tumor proliferation in mouse xenografts. Additionally, HNO inhibits tumor angiogenesis and induces cancer cell apoptosis. The effects seen with HNO donors are quite different from NO donors and in some cases are opposite. The chemical nature of HNO explains how HNO and NO, although closely chemically related, act so differently in biochemical systems. This also gives insight into the potential molecular motifs that may be reactive towards HNO and opens up a novel field of pharmacological development.


Antioxidants & Redox Signaling | 2011

The specificity of nitroxyl chemistry is unique among nitrogen oxides in biological systems

Wilmarie Flores-Santana; Debra J. Salmon; Sonia Donzelli; Christopher H. Switzer; Debashree Basudhar; Lisa A. Ridnour; Robert Y.S. Cheng; Sharon A. Glynn; Nazareno Paolocci; Jon M. Fukuto; Katrina M. Miranda; David A. Wink

The importance of nitric oxide in mammalian physiology has been known for nearly 30 years. Similar attention for other nitrogen oxides such as nitroxyl (HNO) has been more recent. While there has been speculation as to the biosynthesis of HNO, its pharmacological benefits have been demonstrated in several pathophysiological settings such as cardiovascular disorders, cancer, and alcoholism. The chemical biology of HNO has been identified as related to, but unique from, that of its redox congener nitric oxide. A summary of these findings as well as a discussion of possible endogenous sources of HNO is presented in this review.


Journal of Medicinal Chemistry | 2013

Synthesis and Chemical and Biological Comparison of Nitroxyl- and Nitric Oxide-Releasing Diazeniumdiolate-Based Aspirin Derivatives

Debashree Basudhar; Gaurav Bharadwaj; Robert Y.S. Cheng; Sarthak Jain; Sa Shi; Julie Heinecke; Ryan J. Holland; Lisa A. Ridnour; Viviane Menezes Caceres; Regina Celia Spadari-Bratfisch; Nazareno Paolocci; Carlos A. Velázquez-Martínez; David A. Wink; Katrina M. Miranda

Structural modifications of nonsteroidal anti-inflammatory drugs (NSAIDs) have successfully reduced the side effect of gastrointestinal ulceration without affecting anti-inflammatory activity, but they may increase the risk of myocardial infarction with chronic use. The fact that nitroxyl (HNO) reduces platelet aggregation, preconditions against myocardial infarction, and enhances contractility led us to synthesize a diazeniumdiolate-based HNO-releasing aspirin and to compare it to an NO-releasing analogue. Here, the decomposition mechanisms are described for these compounds. In addition to protection against stomach ulceration, these prodrugs exhibited significantly enhanced cytotoxcity compared to either aspirin or the parent diazeniumdiolate toward nonsmall cell lung carcinoma cells (A549), but they were not appreciably toxic toward endothelial cells (HUVECs). The HNO-NSAID prodrug inhibited cylcooxgenase-2 and glyceraldehyde 3-phosphate dehydrogenase activity and triggered significant sarcomere shortening on murine ventricular myocytes compared to control. Together, these anti-inflammatory, antineoplasic, and contractile properties suggest the potential of HNO-NSAIDs in the treatment of inflammation, cancer, or heart failure.


Archives of Pharmacal Research | 2009

Comparing the chemical biology of NO and HNO.

Wilmarie Flores-Santana; Christopher H. Switzer; Lisa A. Ridnour; Debashree Basudhar; Daniele Mancardi; Sonia Donzelli; Douglas D. Thomas; Katrina M. Miranda; Jon M. Fukuto; David A. Wink

For the past couple of decades nitric oxide (NO) and nitroxyl (HNO) have been extensively studied due to the important role they play in many physiological and/or pharmacological processes. Many researchers have reported important signaling pathways as well as mechanisms of action of these species, showing direct and indirect effects depending on the environment. Both NO and HNO can react with, among others, metals, proteins, thiols and heme proteins via unique and distinct chemistry leading to improvement of some clinical conditions. Understanding the basic chemistry of NO and HNO and distinguishing their mechanisms of action as well as methods of detection are crucial for understanding the current and potential clinical applications. In this review, we summarize some of the most important findings regarding NO and HNO chemistry, revealing some of the possible mechanisms of their beneficial actions.


Cancer Research | 2015

NOS Inhibition Modulates Immune Polarization and Improves Radiation-Induced Tumor Growth Delay.

Lisa A. Ridnour; Robert Y.S. Cheng; Jonathan M. Weiss; Sukhbir Kaur; David R. Soto-Pantoja; Debashree Basudhar; Julie Heinecke; C. Andrew Stewart; William DeGraff; Anastasia L. Sowers; Angela Thetford; Aparna H. Kesarwala; David D. Roberts; Howard A. Young; James B. Mitchell; Giorgio Trinchieri; Robert H. Wiltrout; David A. Wink

Nitric oxide synthases (NOS) are important mediators of progrowth signaling in tumor cells, as they regulate angiogenesis, immune response, and immune-mediated wound healing. Ionizing radiation (IR) is also an immune modulator and inducer of wound response. We hypothesized that radiation therapeutic efficacy could be improved by targeting NOS following tumor irradiation. Herein, we show enhanced radiation-induced (10 Gy) tumor growth delay in a syngeneic model (C3H) but not immunosuppressed (Nu/Nu) squamous cell carcinoma tumor-bearing mice treated post-IR with the constitutive NOS inhibitor N(G)-nitro-l-arginine methyl ester (L-NAME). These results suggest a requirement of T cells for improved radiation tumor response. In support of this observation, tumor irradiation induced a rapid increase in the immunosuppressive Th2 cytokine IL10, which was abated by post-IR administration of L-NAME. In vivo suppression of IL10 using an antisense IL10 morpholino also extended the tumor growth delay induced by radiation in a manner similar to L-NAME. Further examination of this mechanism in cultured Jurkat T cells revealed L-NAME suppression of IR-induced IL10 expression, which reaccumulated in the presence of exogenous NO donor. In addition to L-NAME, the guanylyl cyclase inhibitors ODQ and thrombospondin-1 also abated IR-induced IL10 expression in Jurkat T cells and ANA-1 macrophages, which further suggests that the immunosuppressive effects involve eNOS. Moreover, cytotoxic Th1 cytokines, including IL2, IL12p40, and IFNγ, as well as activated CD8(+) T cells were elevated in tumors receiving post-IR L-NAME. Together, these results suggest that post-IR NOS inhibition improves radiation tumor response via Th1 immune polarization within the tumor microenvironment.


Nitric Oxide | 2014

Analysis of the HNO and NO donating properties of alicyclic amine diazeniumdiolates.

Gaurav Bharadwaj; Patricia Graça Zanichelli Benini; Debashree Basudhar; Cyf Ramos-Colon; Gail M. Johnson; Marti Larriva; Larry K. Keefer; Daniela Andrei; Katrina M. Miranda

Nitroxyl (HNO) donors have been shown to elicit a variety of pharmacological responses, ranging from tumoricidal effects to treatment of heart failure. Isopropylamine-based diazeniumdiolates have been shown to produce HNO on decomposition under physiological conditions. Herein, we report the synthesis and HNO release profiles of primary alicyclic amine-based diazeniumdiolates. These compounds extend the range of known diazeniumdiolate-based HNO donors. Acetoxymethyl ester-protected diazeniumdiolates were also synthesized to improve purification and cellular uptake. The acetoxymethyl derivative of cyclopentylamine diazeniumdiolate not only showed higher cytotoxicity toward cancer cells as compared to the parent anion but was also effective in combination with tamoxifen for targeting estrogen receptor α-negative breast cancer cells.


Free Radical Biology and Medicine | 2014

Chemotherapeutic Potential of Diazeniumdiolate-based Aspirin Prodrugs in Breast Cancer

Debashree Basudhar; Robert C. Cheng; Gaurav Bharadwaj; Lisa A. Ridnour; David A. Wink; Katrina M. Miranda

Diazeniumdiolate-based aspirin prodrugs have previously been shown to retain the anti-inflammatory properties of aspirin while protecting against the common side effect of stomach ulceration. Initial analysis of two new prodrugs of aspirin that also release either nitroxyl (HNO) or nitric oxide (NO) demonstrated increased cytotoxicity toward human lung carcinoma cells compared to either aspirin or the parent nitrogen oxide donor. In addition, cytotoxicity was significantly lower in endothelial cells, suggesting cancer-specific sensitivity. To assess the chemotherapeutic potential of these new prodrugs in treatment of breast cancer, we studied their effect both in cultured cells and in a nude mouse model. Both prodrugs reduced growth of breast adenocarcinoma cells more effectively than the parent compounds while not being appreciably cytotoxic in a related nontumorigenic cell line (MCF-10A). The HNO donor also was more cytotoxic than the related NO donor. The basis for the observed specificity was investigated in terms of impact on metabolism, DNA damage and repair, apoptosis, angiogenesis and metastasis. The results suggest a significant pharmacological potential for treatment of breast cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Coexpression of NOS2 and COX2 accelerates tumor growth and reduces survival in estrogen receptor-negative breast cancer

Debashree Basudhar; Sharon A. Glynn; Madison Greer; Veena Somasundaram; Jae Hong No; David A. Scheiblin; Pablo Garrido; William F. Heinz; Aideen E. Ryan; Jonathan M. Weiss; Robert Y.S. Cheng; Lisa A. Ridnour; Stephen Lockett; Daniel W. McVicar; Stefan Ambs; David A. Wink

Significance Nitric oxide synthase-2 (NOS2) and cyclooxygenase-2 (COX2) are inflammation-associated enzymes with oncogenic function in breast cancer. We show that crosstalk between NOS2/COX2 promotes aggressive phenotypes and that elevated coexpression of NOS2/COX2 in tumors predict significantly reduced patient survival (33%) when compared with 95% survival of estrogen receptor-negative patients with low NOS2/COX2 tumor expression. In addition, we have identified a tumor subtype-specific mechanism showing involvement of TNFα and/or endoplasmic reticulum stress as key players in this autocrine loop. Importantly, the simultaneous inhibition of NOS2/COX2 significantly reduced tumor growth in a xenograft murine model, suggesting that targeted inhibition of NOS2/COX2 may be therapeutically beneficial. Proinflammatory signaling pathways are commonly up-regulated in breast cancer. In estrogen receptor-negative (ER−) and triple-negative breast cancer (TNBC), nitric oxide synthase-2 (NOS2) and cyclooxygenase-2 (COX2) have been described as independent predictors of disease outcome. We further explore these findings by investigating the impact of their coexpression on breast cancer survival. Elevated coexpression of NOS2/COX2 proteins is a strong predictor of poor survival among ER− patients (hazard ratio: 21). Furthermore, we found that the key products of NOS2 and COX2, NO and prostaglandin E2 (PGE2), respectively, promote feed-forward NOS2/COX2 crosstalk in both MDA-MB-468 (basal-like) and MDA-MB-231 (mesenchymal-like) TNBC cell lines in which NO induced COX2 and PGE2 induced NOS2 proteins. COX2 induction by NO involved TRAF2 activation that occurred in a TNFα-dependent manner in MDA-MB-468 cells. In contrast, NO-mediated TRAF2 activation in the more aggressive MDA-MB-231 cells was TNFα independent but involved the endoplasmic reticulum stress response. Inhibition of NOS2 and COX2 using amino-guanidine and aspirin/indomethacin yielded an additive reduction in the growth of MDA-MB-231 tumor xenografts. These findings support a role of NOS2/COX2 crosstalk during disease progression of aggressive cancer phenotypes and offer insight into therapeutic applications for better survival of patients with ER− and TNBC disease.


Nitric Oxide | 2014

Gene expression profiles of NO- and HNO-donor treated breast cancer cells: insights into tumor response and resistance pathways.

Robert Y.S. Cheng; Debashree Basudhar; Lisa A. Ridnour; Julie Heinecke; Aparna H. Kesarwala; Sharon A. Glynn; Christopher H. Switzer; Stefan Ambs; Katrina M. Miranda; David A. Wink

Nitric oxide (NO) synthase 2 (NOS2), a major inflammatory protein, modulates disease progression via NO in a number of pathologies, including cancer. The role of NOS2-derived NO is not only flux-dependent, which is higher in mouse vs human cells, but also varies based on spatial and temporal distribution both within tumor cells and in the tumor microenvironment. NO donors have been utilized to mimic NO flux conditions and to investigate the effects of varied NO concentrations. As a wide range of effects mediated by NO and other nitrogen oxides such as nitroxyl (HNO) have been elucidated, multiple NO- and HNO-releasing compounds have been developed as potential therapeutics, including as tumor modulators. One of the challenges is to determine differences in biomarker expression from extracellular vs intracellular generation of NO or HNO. Taking advantage of new NO and HNO releasing agents, we have characterized the gene expression profile of estrogen receptor-negative human breast cancer (MDA-MB-231) cells following exposure to aspirin, the NO donor DEA/NO, the HNO donor IPA/NO andtheir intracellularly-activated prodrug conjugates DEA/NO-aspirin and IPA/NO-aspirin. Comparison of the gene expression profiles demonstrated that several genes were uniquely expressed with respect to NO or HNO, such as miR-21, HSP70, cystathionine γ-lyase and IL24. These findings provide insight into targets and pathways that could be therapeutically exploited by the redox related species NO and HNO.


British Journal of Pharmacology | 2018

Understanding the tumour microenvironment communication network from a NOS2/COX2 perspective

Debashree Basudhar; Gaurav Bharadwaj; Veena Somasundaram; Robert Y.S. Cheng; Lisa A. Ridnour; Mayumi Fujita; Stephen Lockett; Stephen K. Anderson; Daniel W. McVicar; David A. Wink

Recent findings suggest that co‐expression of NOS2 and COX2 is a strong prognostic indicator in triple‐negative breast cancer patients. These two key inflammation‐associated enzymes are responsible for the biosynthesis of NO and PGE2, respectively, and can exert their effect in both an autocrine and paracrine manner. Impairment of their physiological regulation leads to critical changes in both intra‐tumoural and intercellular communication with the immune system and their adaptation to the hypoxic tumour micro‐environment. Recent studies have also established a key role of NOS2–COX2 in causing metabolic shift. This review provides an extensive overview of the role of NO and PGE2 in shaping communication between the tumour micro‐environment composed of tumour and immune cells that in turn favours tumour progression and metastasis.

Collaboration


Dive into the Debashree Basudhar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lisa A. Ridnour

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert Y.S. Cheng

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Veena Somasundaram

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel W. McVicar

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gaurav Bharadwaj

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Julie Heinecke

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sharon A. Glynn

National University of Ireland

View shared research outputs
Researchain Logo
Decentralizing Knowledge