Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
JCI Insight ; 9(12)2024 May 21.
Article in English | MEDLINE | ID: mdl-38912586

ABSTRACT

Immune therapy is the new frontier of cancer treatment. Therapeutic radiation is a known inducer of immune response and can be limited by immunosuppressive mediators including cyclooxygenase-2 (COX2) that is highly expressed in aggressive triple negative breast cancer (TNBC). A clinical cohort of TNBC tumors revealed poor radiation therapeutic efficacy in tumors expressing high COX2. Herein, we show that radiation combined with adjuvant NSAID (indomethacin) treatment provides a powerful combination to reduce both primary tumor growth and lung metastasis in aggressive 4T1 TNBC tumors, which occurs in part through increased antitumor immune response. Spatial immunological changes including augmented lymphoid infiltration into the tumor epithelium and locally increased cGAS/STING1 and type I IFN gene expression were observed in radiation-indomethacin-treated 4T1 tumors. Thus, radiation and adjuvant NSAID treatment shifts "immune desert phenotypes" toward antitumor M1/TH1 immune mediators in these immunologically challenging tumors. Importantly, radiation-indomethacin combination treatment improved local control of the primary lesion, reduced metastatic burden, and increased median survival when compared with radiation treatment alone. These results show that clinically available NSAIDs can improve radiation therapeutic efficacy through increased antitumor immune response and augmented local generation of cGAS/STING1 and type I IFNs.


Subject(s)
Membrane Proteins , Signal Transduction , T-Lymphocytes, Cytotoxic , Animals , Membrane Proteins/metabolism , Mice , Female , Signal Transduction/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/radiotherapy , Indomethacin/pharmacology , Indomethacin/therapeutic use , Cell Line, Tumor , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Nucleotidyltransferases/metabolism , Interferon Type I/metabolism , Cyclooxygenase 2/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Mice, Inbred BALB C
2.
J Vis Exp ; (157)2020 03 20.
Article in English | MEDLINE | ID: mdl-32250353

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with limited therapeutic options. When compared to patients with less aggressive breast tumors, the 5-year survival rate of TNBC patients is 77% due to their characteristic drug-resistant phenotype and metastatic burden. Toward this end, murine models have been established aimed at identifying novel therapeutic strategies limiting TNBC tumor growth and metastatic spread. This work describes a practical guide for the TNBC orthotopic model where MDA-MB-231 breast cancer cells suspended in a basement membrane matrix are implanted in the fourth mammary fat pad, which closely mimics the cancer cell behavior in humans. Measurement of tumors by caliper, lung metastasis assessment via in vivo and ex vivo imaging, and molecular detection are discussed. This model provides an excellent platform to study therapeutic efficacy and is especially suitable for the study of the interaction between the primary tumor and distal metastatic sites.


Subject(s)
Triple Negative Breast Neoplasms/pathology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Lung Neoplasms/secondary , Mice , Phenotype , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
3.
Redox Biol ; 28: 101354, 2020 01.
Article in English | MEDLINE | ID: mdl-31683257

ABSTRACT

The role of nitric oxide (NO) in cancer progression has largely been studied in the context of tumor NOS2 expression. However, pro- versus anti-tumor signaling is also affected by tumor cell-macrophage interactions. While these cell-cell interactions are partly regulated by NO, the functional effects of NO flux on proinflammatory (M1) macrophages are unknown. Using a triple negative murine breast cancer model, we explored the potential role of macrophage Nos2 on 4T1 tumor progression. The effects of NO on macrophage phenotype were examined in bone marrow derived macrophages from wild type and Nos2-/- mice following in vitro stimulation with cytokine/LPS combinations to produce low, medium, and high NO flux. Remarkably, Nos2 induction was spatially distinct, where Nos2high cells expressed low cyclooxygenase-2 (Cox2) and vice versa. Importantly, in vitro M1 polarization with IFNγ+LPS induced high NO flux that was restricted to cells harboring depolarized mitochondria. This flux altered the magnitude and spatial extent of hypoxic gradients. Metabolic and single cell analyses demonstrated that single cell Nos2 induction limited the generation of hypoxic gradients in vitro, and Nos2-dependent and independent features may collaborate to regulate M1 functionality. It was found that Cox2 expression was important for Nos2high cells to maintain NO tolerance. Furthermore, Nos2 and Cox2 expression in 4T1 mouse tumors was spatially orthogonal forming distinct cellular neighborhoods. In summary, the location and type of Nos2high cells, NO flux, and the inflammatory status of other cells, such as Cox2high cells in the tumor niche contribute to Nos2 inflammatory mechanisms that promote disease progression of 4T1 tumors.


Subject(s)
Cytokines/metabolism , Lipopolysaccharides/adverse effects , Nitric Oxide Synthase Type II/genetics , Triple Negative Breast Neoplasms/pathology , Animals , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , Macrophages/drug effects , Macrophages/immunology , Mice , Neoplasm Transplantation , Nitric Oxide/metabolism , Single-Cell Analysis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
4.
Redox Biol ; 22: 101158, 2019 04.
Article in English | MEDLINE | ID: mdl-30852389

ABSTRACT

Pancreatic cancer is a highly metastatic tumor with an extremely low 5-year survival rate. Lack of efficient diagnostics and dearth of effective therapeutics that can target the cancer as well as the microenvironment niche are the reasons for limited success in treatment and management of this disease. Cell invasion through extracellular matrix (ECM) involves the complex regulation of adhesion to and detachment from ECM and its understanding is critical to metastatic potential of pancreatic cancer. To understand the characteristics of these cancer cells and their ability to metastasize, we compared human pancreatic cancer cell line, PANC-1 and its invading phenotype (INV) collected from transwell inserts. The invasive cell type, INV, exhibited higher resistance to Carbon-ion radiation compared to whole cultured (normally dish-cultured) PANC-1 (WCC), and had more efficient in vitro spheroid formation capability. Invasiveness of INV was hampered by nitric oxide synthase (NOS) inhibitors, suggesting that nitric oxide (NO) plays a cardinal role in PANC-1 invasion. In addition, in vitro studies indicated that a MEK-ERK-dependent, JAK independent mechanism through which NOS/NO modulate PANC-1 invasiveness. Suspended INV showed enhanced NO production as well as induction of several pro-metastatic, and stemness-related genes. NOS inhibitor, l-NAME, reduced the expression of these pro-metastatic or stemness-related genes, and dampened spheroid formation ability, suggesting that NO can potentially influence pancreatic cancer aggressiveness. Furthermore, xenograft studies with INV and WCC in NSG mouse model revealed a greater ability of INV compared to WCC, to metastasize to the liver and l-NAME diminished the metastatic lesions in mice injected with INV. Overall, data suggest that NO is a key player associated with resistance to radiation and metastasis of pancreatic cancer; and inhibition of NOS demonstrates therapeutic potential as observed in the animal model by specifically targeting the metastatic cells that harbor stem-like features and are potentially responsible for relapse.


Subject(s)
Nitric Oxide/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phenotype , Animals , Cell Line, Tumor , Disease Models, Animal , Fluorescent Antibody Technique , Humans , MAP Kinase Signaling System , Male , Mice , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Nitric Oxide Synthase/metabolism
5.
Antioxid Redox Signal ; 30(8): 1124-1143, 2019 03 10.
Article in English | MEDLINE | ID: mdl-29634348

ABSTRACT

SIGNIFICANCE: Cancer is a complex disease, which not only involves the tumor but its microenvironment comprising different immune cells as well. Nitric oxide (NO) plays specific roles within tumor cells and the microenvironment and determines the rate of cancer progression, therapy efficacy, and patient prognosis. Recent Advances: Key understanding of the processes leading to dysregulated NO flux within the tumor microenvironment over the past decade has provided better understanding of the dichotomous role of NO in cancer and its importance in shaping the immune landscape. It is becoming increasingly evident that nitric oxide synthase 2 (NOS2)-mediated NO/reactive nitrogen oxide species (RNS) are heavily involved in cancer progression and metastasis in different types of tumor. More recent studies have found that NO from NOS2+ macrophages is required for cancer immunotherapy to be effective. CRITICAL ISSUES: NO/RNS, unlike other molecules, are unique in their ability to target a plethora of oncogenic pathways during cancer progression. In this review, we subcategorize the different levels of NO produced by cells and shed light on the context-dependent temporal effects on cancer signaling and metabolic shift in the tumor microenvironment. FUTURE DIRECTIONS: Understanding the source of NO and its spaciotemporal profile within the tumor microenvironment could help improve efficacy of cancer immunotherapies by improving tumor infiltration of immune cells for better tumor clearance.


Subject(s)
Neoplasms/metabolism , Nitric Oxide/metabolism , Reactive Nitrogen Species/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Nitric Oxide Synthase Type II/metabolism , Prognosis , Signal Transduction , Tumor Microenvironment
6.
Br J Pharmacol ; 176(2): 155-176, 2019 01.
Article in English | MEDLINE | ID: mdl-30152521

ABSTRACT

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. LINKED ARTICLES: This article is part of a themed section on Nitric Oxide 20 Years from the 1998 Nobel Prize. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.2/issuetoc.


Subject(s)
Cyclooxygenase 2/metabolism , Nitric Oxide Synthase Type II/metabolism , Animals , Humans , Nitric Oxide/metabolism , Tumor Microenvironment
7.
Proc Natl Acad Sci U S A ; 114(49): 13030-13035, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29087320

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Cyclooxygenase 2/genetics , Gene Expression Regulation, Neoplastic , Nitric Oxide Synthase Type II/genetics , Receptors, Estrogen/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Aspirin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Endoplasmic Reticulum Stress/drug effects , Feedback, Physiological , Female , Guanidines/pharmacology , Humans , Indomethacin/pharmacology , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Prognosis , Proportional Hazards Models , Receptors, Estrogen/deficiency , Signal Transduction , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
8.
Antioxid Redox Signal ; 26(18): 1044-1058, 2017 06 20.
Article in English | MEDLINE | ID: mdl-27464521

ABSTRACT

SIGNIFICANCE: Breast cancer is the second leading cause of cancer-related deaths among women in the United States. Development and progression of malignancy are associated with diverse cell signaling pathways that control cell proliferation, survival, motility, invasion, and metastasis. Recent Advances: An increasing number of clinical studies have implicated a strong relationship between elevated tumor nitric oxide synthase-2 (NOS2) expression and poor patient survival. CRITICAL ISSUES: Herein, we review what we believe to be key mechanisms in the role(s) of NOS2-derived nitric oxide (NO) as a driver of breast cancer disease progression. High NO increases cyclooxygenase-2 activity, hypoxia inducible factor-1 alpha protein stabilization, and activation of important cell signaling pathways, including phosphoinositide 3-kinase/protein kinase B, mitogen-activated protein kinase, epidermal growth factor receptor, and Ras, through post-translational protein modifications. Moreover, dysregulated NO flux within the tumor microenvironment has other important roles, including the promotion of angiogenesis and modulation of matrix metalloproteinase/tissue inhibitor matrix metalloproteinase associated with tumor progression. FUTURE DIRECTIONS: The elucidation of these and other NO-driven pathways implicates NOS2 as a key driver of breast cancer disease progression and provides a new perspective in the identification of novel targets that may be therapeutically beneficial in the treatment of estrogen receptor-negative disease. Antioxid. Redox Signal. 26, 1044-1058.


Subject(s)
Breast Neoplasms/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Animals , Disease Progression , Female , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein Stability , Signal Transduction , Tumor Microenvironment
9.
Antioxid Redox Signal ; 26(18): 1059-1077, 2017 06 20.
Article in English | MEDLINE | ID: mdl-27494631

ABSTRACT

SIGNIFICANCE: Gastrointestinal (GI) cancer taken together constitutes one of the most common cancers worldwide with a broad range of etiological mechanisms. In this review, we have examined the impact of nitric oxide (NO) on the etiology of colon, colorectal, gastric, esophageal, and liver cancers. Recent Advances: Despite differences in etiology, initiation, and progression, chronic inflammation has been shown to be a common element within these cancers showing interactions of numerous pathways. NO generated at the inflammatory site contributes to the initiation and progression of disease. The amount of NO generated, time, and site vary and are an important determinant of the biological effects initiated. Among the nitric oxide synthase enzymes, the inducible isoform has the most diverse range, participating in numerous carcinogenic processes. There is emerging evidence showing that inducible nitric oxide synthase (NOS2) plays a central role in the process of tumor initiation and/or development. CRITICAL ISSUES: Redox inflammation through NOS2 and cyclooxygenase-2 participates in driving the mechanisms of initiation and progression in GI cancers. FUTURE DIRECTIONS: Understanding the underlying mechanism involved in NOS2 activation can provide new insights into important prevention and treatment strategies. Antioxid. Redox Signal. 26, 1059-1077.


Subject(s)
Cyclooxygenase 2/metabolism , Gastrointestinal Neoplasms/enzymology , Inflammation/enzymology , Nitric Oxide Synthase Type II/metabolism , Animals , Carcinogenesis/metabolism , Disease Progression , Gastrointestinal Neoplasms/etiology , Humans , Inflammation/etiology , Nitric Oxide/immunology , Oxidation-Reduction
10.
J Biol Chem ; 291(31): 16100-11, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27235395

ABSTRACT

Mycobacterium tuberculosis DosS is critical for the induction of M. tuberculosis dormancy genes in response to nitric oxide (NO), carbon monoxide (CO), or hypoxia. These environmental stimuli, which are sensed by the DosS heme group, result in autophosphorylation of a DosS His residue, followed by phosphotransfer to an Asp residue of the response regulator DosR. To clarify the mechanism of gaseous ligand recognition and signaling, we investigated the hydrogen-bonding interactions of the iron-bound CO and NO ligands by site-directed mutagenesis of Glu-87 and His-89. Autophosphorylation assays and molecular dynamics simulations suggest that Glu-87 has an important role in ligand recognition, whereas His-89 is essential for signal transduction to the kinase domain, a process for which Arg-204 is important. Mutation of Glu-87 to Ala or Gly rendered the protein constitutively active as a kinase, but with lower autophosphorylation activity than the wild-type in the Fe(II) and the Fe(II)-CO states, whereas the E87D mutant had little kinase activity except for the Fe(II)-NO complex. The H89R mutant exhibited attenuated autophosphorylation activity, although the H89A and R204A mutants were inactive as kinases, emphasizing the importance of these residues in communication to the kinase core. Resonance Raman spectroscopy of the wild-type and H89A mutant indicates the mutation does not alter the heme coordination number, spin state, or porphyrin deformation state, but it suggests that interdomain interactions are disrupted by the mutation. Overall, these results confirm the importance of the distal hydrogen-bonding network in ligand recognition and communication to the kinase domain and reveal the sensitivity of the system to subtle differences in the binding of gaseous ligands.


Subject(s)
Bacterial Proteins , Carbon Monoxide , Mycobacterium tuberculosis , Nitric Oxide , Protamine Kinase , Signal Transduction/physiology , Amino Acid Substitution , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbon Monoxide/chemistry , Carbon Monoxide/metabolism , Hydrogen Bonding , Mutation, Missense , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Phosphorylation , Protamine Kinase/chemistry , Protamine Kinase/genetics , Protamine Kinase/metabolism
11.
J Biol Chem ; 291(14): 7325-33, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26833565

ABSTRACT

Cholest-4-en-3-one, whether added exogenously or generated intracellularly from cholesterol, inhibits the growth ofMycobacterium tuberculosiswhen CYP125A1 and CYP142A1, the cytochrome P450 enzymes that initiate degradation of the sterol side chain, are disabled. Here we demonstrate that a 16-hydroxy derivative of cholesterol, which was previously reported to inhibit growth ofM. tuberculosis, acts by preventing the oxidation of the sterol side chain even in the presence of the relevant cytochrome P450 enzymes. The finding that (25R)-cholest-5-en-3ß,16ß,26-triol (1) (and its 3-keto metabolite) inhibit growth suggests that cholesterol analogs with non-degradable side chains represent a novel class of anti-mycobacterial agents. In accord with this, two cholesterol analogs with truncated, fluorinated side chains have been synthesized and shown to similarly block the growth in culture ofM. tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Cholestenones/pharmacology , Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Mycobacterium tuberculosis/growth & development , Antitubercular Agents/chemistry , Bacterial Proteins/metabolism , Cholestenones/chemistry , Cholesterol/chemistry , Cytochrome P-450 Enzyme System/metabolism
12.
Coord Chem Rev ; 306(Pt 2): 708-723, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26688591

ABSTRACT

Small redox active molecules such as reactive nitrogen and oxygen species and hydrogen sulfide have emerged as important biological mediators that are involved in various physiological and pathophysiological processes. Advancement in understanding of cellular mechanisms that tightly regulate both generation and reactivity of these molecules is central to improved management of various disease states including cancer and cardiovascular dysfunction. Imbalance in the production of redox active molecules can lead to damage of critical cellular components such as cell membranes, proteins and DNA and thus may trigger the onset of disease. These small inorganic molecules react independently as well as in a concerted manner to mediate physiological responses. This review provides a general overview of the redox biology of these key molecules, their diverse chemistry relevant to physiological processes and their interrelated nature in cellular signaling.

13.
Cancer Res ; 75(14): 2788-99, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25990221

ABSTRACT

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.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Enzyme Inhibitors/pharmacology , Lymphocyte Activation/drug effects , Neoplasms/pathology , Nitric Oxide Synthase/antagonists & inhibitors , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Enzyme Inhibitors/therapeutic use , Female , Humans , Jurkat Cells , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasms/drug therapy , Neoplasms/radiotherapy
14.
Free Radic Biol Med ; 83: 101-14, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25659932

ABSTRACT

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.


Subject(s)
Adenocarcinoma/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Aspirin/pharmacology , Azo Compounds/chemistry , Breast Neoplasms/drug therapy , Prodrugs/pharmacology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Aspirin/chemistry , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , Female , Humans , Mice , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitrogen Oxides/metabolism , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
J Biol Chem ; 290(16): 10000-17, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25670859

ABSTRACT

Defining the conformational states of cytochrome P450 active sites is critical for the design of agents that minimize drug-drug interactions, the development of isoform-specific P450 inhibitors, and the engineering of novel oxidative catalysts. We used two-dimensional (1)H,(15)N HSQC chemical shift perturbation mapping of (15)N-labeled Phe residues and x-ray crystallography to examine the ligand-dependent conformational dynamics of CYP119. Active site Phe residues were most affected by the binding of azole inhibitors and fatty acid substrates, in agreement with active site localization of the conformational changes. This was supported by crystallography, which revealed movement of the F-G loop with various azoles. Nevertheless, the NMR chemical shift perturbations caused by azoles and substrates were distinguishable. The absence of significant chemical shift perturbations with several azoles revealed binding of ligands to an open conformation similar to that of the ligand-free state. In contrast, 4-phenylimidazole caused pronounced NMR changes involving Phe-87, Phe-144, and Phe-153 that support the closed conformation found in the crystal structure. The same closed conformation is observed by NMR and crystallography with a para-fluoro substituent on the 4-phenylimidazole, but a para-chloro or bromo substituent engendered a second closed conformation. An open conformation is thus favored in solution with many azole ligands, but para-substituted phenylimidazoles give rise to two closed conformations that depend on the size of the para-substituent. The results suggest that ligands selectively stabilize discrete cytochrome P450 conformational states.


Subject(s)
Archaeal Proteins/chemistry , Cytochrome P-450 Enzyme System/chemistry , Models, Molecular , Sulfolobus acidocaldarius/chemistry , Amino Acid Motifs , Archaeal Proteins/antagonists & inhibitors , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/chemistry , Fatty Acids/chemistry , Gene Expression , Imidazoles/chemistry , Ligands , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptide Mapping , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sulfolobus acidocaldarius/enzymology
16.
Nitric Oxide ; 42: 70-8, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25192820

ABSTRACT

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.


Subject(s)
Azo Compounds/chemistry , Nitric Oxide/metabolism , Magnetic Resonance Spectroscopy , Spectrophotometry, Ultraviolet
17.
Nitric Oxide ; 43: 17-28, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25153034

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide/pharmacology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Profiling , Humans , Mice , Nitric Oxide Synthase/metabolism , Signal Transduction
18.
J Med Chem ; 56(20): 7804-20, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24102516

ABSTRACT

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.


Subject(s)
Aspirin/chemical synthesis , Aspirin/pharmacology , Azo Compounds/chemistry , Nitric Oxide/chemistry , Nitrogen Oxides/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/chemistry , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Cyclooxygenase 2/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/metabolism , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Models, Chemical , Molecular Structure , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/pharmacology , Sarcomeres/drug effects , Sarcomeres/metabolism
19.
Antioxid Redox Signal ; 14(9): 1659-74, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21235346

ABSTRACT

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.


Subject(s)
Nitrogen Oxides/metabolism , Animals , Humans , Metalloproteins/chemistry , Metalloproteins/metabolism , Nitrogen Oxides/chemistry , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism
20.
Arch Pharm Res ; 32(8): 1139-53, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19727606

ABSTRACT

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.


Subject(s)
Nitric Oxide/chemistry , Nitrogen Oxides/chemistry , Animals , Humans , Nitric Oxide/physiology , Nitrogen Oxides/metabolism , Reactive Nitrogen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...