Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
Mol Oncol ; 17(12): 2526-2545, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37149844

ABSTRACT

Synthetic oleanane triterpenoids (SOTs) are small molecules with broad anticancer properties. A recently developed SOT, 1-[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]-4(-pyridin-2-yl)-1H-imidazole (CDDO-2P-Im or '2P-Im'), exhibits enhanced activity and improved pharmacokinetics over CDDO-Im, a previous generation SOT. However, the mechanisms leading to these properties are not defined. Here, we show the synergy of 2P-Im and the proteasome inhibitor ixazomib in human multiple myeloma (MM) cells and 2P-Im activity in a murine model of plasmacytoma. RNA sequencing and quantitative reverse transcription PCR revealed the upregulation of the unfolded protein response (UPR) in MM cells upon 2P-lm treatment, implicating the activation of the UPR as a key step in 2P-Im-induced apoptosis. Supporting this hypothesis, the deletion of genes encoding either protein kinase R-like endoplasmic reticulum kinase (PERK) or DNA damage-inducible transcript 3 protein (DDIT3; also known as CHOP) impaired the MM response to 2P-Im, as did treatment with ISRIB, integrated stress response inhibitor, which inhibits UPR signaling downstream of PERK. Finally, both drug affinity responsive target stability and thermal shift assays demonstrated direct binding of 2P-Im to endoplasmic reticulum chaperone BiP (GRP78/BiP), a stress-inducible key signaling molecule of the UPR. These data reveal GRP78/BiP as a novel target of SOTs, and specifically of 2P-Im, and suggest the potential broader utility of this class of small molecules as modulators of the UPR.


Subject(s)
Multiple Myeloma , Humans , Mice , Animals , Multiple Myeloma/drug therapy , Endoplasmic Reticulum Chaperone BiP , Cell Line, Tumor , Apoptosis , Imidazoles/pharmacology , Unfolded Protein Response
2.
Free Radic Biol Med ; 188: 206-220, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35753588

ABSTRACT

Radiation-induced oral mucositis is one of the most common adverse events in radiation therapy for head and neck cancers, but treatments for oral mucositis are limited to palliative and supportive care. New approaches are required to prevent radiation-induced mucositis and to improve treatments. The Keap1-Nrf2 system regulates cytoprotection against oxidative and electrophilic stresses. Nrf2 also regulates keratin layer thickness in mouse tongues. Therefore, we hypothesized that Nrf2 may protect the tongue epithelium against radiation-induced mucositis via elimination of reactive oxygen species and induction of keratin layer thickening. To test this hypothesis, we prepared a system for γ-ray exposure of restricted areas and irradiated the tongues of model mice with Nrf2 and Keap1 loss-of-function. We discovered that loss of Nrf2 expression indeed sensitized the tongue epithelium to radiation-induced ulcer formation with inflammation. Constitutive Nrf2 activation by genetic Keap1 knockdown alleviated radiation-induced DNA damage by increasing antioxidation. In agreement with the genetic Nrf2 activation model, the Nrf2 inducer CDDO-Im prevented irradiation damage to the tongue epithelium. These results demonstrate that Nrf2 activation has the potential to prevent the development of radiation-induced mucositis and that Nrf2 inducers are an important therapeutic drug for protection of the upper aerodigestive tract from radiation-induced mucositis.


Subject(s)
Mucositis , Radiation Injuries , Stomatitis , Animals , Mice , Antioxidants/pharmacology , Cytoskeletal Proteins/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Keratins/metabolism , Mucositis/etiology , Mucositis/prevention & control , NF-E2-Related Factor 2/metabolism , Oleanolic Acid/analogs & derivatives , Radiation Injuries/prevention & control , Stomatitis/etiology , Stomatitis/prevention & control
3.
Mol Cell Biol ; 41(11): e0023621, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34398680

ABSTRACT

Heme oxygenase 1 (HO-1) is the key enzyme for heme catabolism and cytoprotection. Whereas HO-1 gene expression in response to various stresses has been investigated extensively, the precise mechanisms by which HO-1 gene expression is regulated by the HO-1 substrate heme remain elusive. To systematically examine whether stress-mediated induction and substrate-mediated induction of HO-1 utilize similar or distinct regulatory pathways, we developed an HO-1-DsRed-knock-in reporter mouse in which the HO-1 gene is floxed by loxP sites and the DsRed gene has been inserted. Myeloid lineage-specific recombination of the floxed locus led to fluorescence derived from expression of the HO-1-DsRed fusion protein in peritoneal macrophages. We also challenged general recombination of the locus and generated mice harboring heterozygous recombinant alleles, which enabled us to monitor HO-1-DsRed expression in the whole body in vivo and ex vivo. HO-1 inducers upregulated HO-1-DsRed expression in myeloid lineage cells isolated from the mice. Notably, analyses of peritoneal macrophages from HO-1-DsRed mice lacking NRF2, a major regulator of the oxidative/electrophilic stress response, led us to identify NRF2-dependent stress response-mediated HO-1 induction and NRF2-independent substrate-mediated HO-1 induction. Thus, the HO-1 gene is subjected to at least two distinct levels of regulation, and the available lines of evidence suggest that substrate induction in peritoneal macrophages is independent of CNC family-based regulation.


Subject(s)
Gene Expression Regulation/genetics , Heme Oxygenase-1/genetics , Heme/metabolism , Macrophages, Peritoneal/metabolism , Membrane Proteins/genetics , Oxidative Stress/genetics , Animals , Antioxidants/metabolism , Bilirubin/metabolism , Biliverdine/metabolism , Heme Oxygenase-1/biosynthesis , Luminescent Proteins/genetics , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Oxidative Stress/physiology
4.
J Med Chem ; 63(17): 9965-9976, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32787104

ABSTRACT

Synthetic triterpenoids including CDDO, its methyl ester (CDDO-Me, bardoxolone methyl), and its imidazolide (CDDO-Im) enhance Nrf2-mediated antioxidant and anti-inflammatory activity in many diseases by reacting with thiols on the adaptor protein, Keap1. Unlike monofunctional CDDO-Me, the bifunctional analog, CDDO-Im, has a second reactive site (imidazolide) and can covalently bind to amino acids other than cysteine on target proteins such as glutathione S-transferase pi (GSTP), serum albumin, or Keap1. Here we show for the first time that bifunctional CDDO-Im (in contrast to CDDO-Me), as low as 50 nM, can covalently transacylate arginine and serine residues in GSTP and cross-link them to adjacent cysteine residues. Moreover, we show that CDDO-Im binds covalently to Keap1 by forming permanent Michael adducts with eight different cysteines, and acyl adducts with lysine and several tyrosine residues. Modeling studies suggest that the Tyr 85 adduct stabilizes the Keap1-Cul3 complex, thereby enhancing the potency of CDDO-Im.


Subject(s)
Imidazoles/chemistry , Kelch-Like ECH-Associated Protein 1/chemistry , Oleanolic Acid/analogs & derivatives , Amino Acid Sequence , Cullin Proteins/chemistry , Cullin Proteins/metabolism , Glutathione S-Transferase pi/chemistry , Glutathione S-Transferase pi/metabolism , Humans , Imidazoles/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Molecular Docking Simulation , Oleanolic Acid/chemistry , Oleanolic Acid/metabolism , Protein Multimerization/drug effects , Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism
5.
NPJ Breast Cancer ; 5: 39, 2019.
Article in English | MEDLINE | ID: mdl-31700995

ABSTRACT

Despite numerous therapeutic advances in the past decade, breast cancer is expected to cause over 42,000 deaths in the United States in 2019. Breast cancer had been considered an immunologically silent tumor; however recent findings suggest that immune cells play important roles in tumor growth even in the breast. Retinoid X receptors (RXRs) are a subclass of nuclear receptors that act as ligand-dependent transcription factors that regulate a variety of cellular processes including proliferation and differentiation; in addition, they are essential for macrophage biology. Rexinoids are synthetic molecules that bind and activate RXRs. Bexarotene is the only rexinoid approved by the FDA for the treatment of refractory cutaneous T-cell lymphoma. Other more-potent rexinoids have been synthesized, such as LG100268 (LG268). Here, we report that treatment with LG 268, but not bexarotene, decreased infiltration of myeloid-derived suppressor cells and CD206-expressing macrophages, increased the expression of PD-L1 by 50%, and increased the ratio of CD8/CD4, CD25 T cells, which correlates with increased cytotoxic activity of CD8 T cells in tumors of MMTV-Neu mice (a model of HER2-positive breast cancer). In the MMTV-PyMT murine model of triple negative breast cancer, LG268 treatment of established tumors prolonged survival, and in combination with anti-PD-L1 antibodies, significantly (p = 0.05) increased the infiltration of cytotoxic CD8 T cells and apoptosis. Collectively, these data suggest that the use of LG268, a RXR agonist, can improve response to immune checkpoint blockade in HER2+ or triple-negative breast cancer.

6.
Cancer Prev Res (Phila) ; 12(4): 211-224, 2019 04.
Article in English | MEDLINE | ID: mdl-30760500

ABSTRACT

Rexinoids, selective ligands for retinoid X receptors (RXR), have shown promise in preventing many types of cancer. However, the limited efficacy and undesirable lipidemic side-effects of the only clinically approved rexinoid, bexarotene, drive the search for new and better rexinoids. Here we report the evaluation of novel pyrimidinyl (Py) analogues of two known chemopreventive rexinoids, bexarotene (Bex) and LG100268 (LG268) in a new paradigm. We show that these novel derivatives were more effective agents than bexarotene for preventing lung carcinogenesis induced by a carcinogen. In addition, these new analogues have an improved safety profile. PyBex caused less elevation of plasma triglyceride levels than bexarotene, while PyLG268 reduced plasma cholesterol levels and hepatomegaly compared with LG100268. Notably, this new paradigm mechanistically emphasizes the immunomodulatory and anti-inflammatory activities of rexinoids. We reveal new immunomodulatory actions of the above rexinoids, especially their ability to diminish the percentage of macrophages and myeloid-derived suppressor cells in the lung and to redirect activation of M2 macrophages. The rexinoids also potently inhibit critical inflammatory mediators including IL6, IL1ß, CCL9, and nitric oxide synthase (iNOS) induced by lipopolysaccharide. Moreover, in vitro iNOS and SREBP (sterol regulatory element-binding protein) induction assays correlate with in vivo efficacy and toxicity, respectively. Our results not only report novel pyrimidine derivatives of existing rexinoids, but also describe a series of biological screening assays that will guide the synthesis of additional rexinoids. Further progress in rexinoid synthesis, potency, and safety should eventually lead to a clinically acceptable and useful new drug for patients with cancer.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Lung Neoplasms/drug therapy , Tetrahydronaphthalenes/pharmacology , Animals , Apoptosis , Bexarotene/pharmacology , Cell Proliferation , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred A , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Cancer Prev Res (Phila) ; 11(3): 143-156, 2018 03.
Article in English | MEDLINE | ID: mdl-29246957

ABSTRACT

Breast cancer and lung cancer remain the top two leading causes of cancer-related deaths in women. Because of limited success in reducing the high mortality of these diseases, new drugs and approaches are desperately needed. Cancer prevention is one such promising strategy that is effective in both preclinical and clinical studies. I-BET 762 is a new bromodomain inhibitor that reversibly targets BET (bromodomain and extraterminal) proteins and impairs their ability to bind to acetylated lysines on histones, thus interrupting downstream transcription. This inhibitor has anti-inflammatory effects and induces growth arrest in many cancers and is currently under clinical trials for treatment of cancer. However, few studies have investigated the chemopreventive effects of bromodomain inhibitors. Here, we found that I-BET 762 significantly delayed tumor development in preclinical breast and lung cancer mouse models. This drug not only induced growth arrest and downregulated c-Myc, pSTAT3, and pERK protein expression in tumor cells in vitro and in vivo but also altered immune populations in different organs. These results demonstrate the promising potential of using I-BET 762 for cancer prevention and suggest the striking effects of I-BET 762 are the result of targeting both tumor cells and the tumor microenvironment. Cancer Prev Res; 11(3); 143-56. ©2017 AACR.


Subject(s)
Benzodiazepines/therapeutic use , Chemoprevention/methods , Lung Neoplasms/prevention & control , Mammary Neoplasms, Experimental/prevention & control , A549 Cells , Animals , Female , Humans , Lung Neoplasms/pathology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred Strains , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Org Biomol Chem ; 15(28): 6001-6005, 2017 Jul 19.
Article in English | MEDLINE | ID: mdl-28678272

ABSTRACT

We report the synthesis and biological activity of C-24 demethyl CDDO-Me 2 and the C-28 amide derivatives 3 and 4, which are analogues of the anti-inflammatory synthetic triterpenoid bardoxolone methyl (CDDO-Me) 1. Demethylation of the C-24 methyl group was accomplished via "abnormal Beckmann" rearrangement and subsequent ring A reformation. Amides 3 and 4 were found to be potent inhibitors of the production of the inflammatory mediator NO in vitro.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drug Design , Inflammation/drug therapy , Oleanolic Acid/analogs & derivatives , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Female , Inflammation/metabolism , Mice , Molecular Conformation , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Oleanolic Acid/chemical synthesis , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , RAW 264.7 Cells
9.
Cancer Lett ; 394: 76-87, 2017 05 28.
Article in English | MEDLINE | ID: mdl-28254412

ABSTRACT

Bromodomain inhibitors (JQ1 and I-BET 762) are a new generation of selective, small molecule inhibitors that target BET (bromodomain and extra terminal) proteins. By impairing their ability to bind to acetylated lysines on histones, bromodomain inhibitors interfere with transcriptional initiation and elongation. BET proteins regulate several genes responsible for cell cycle, apoptosis and inflammation. In this study, JQ1 and I-BET 762 decreased c-Myc and p-Erk 1/2 protein levels and inhibited proliferation in pancreatic cancer cells. The tumor microenvironment is known to play an important role in pancreatic cancer, and these drugs suppressed the production of nitric oxide and a variety of inflammatory cytokines, including IL-6, CCL2, and GM-CSF, in both immune and pancreatic cancer cells in vitro. Notably, the bromodomain inhibitors also reduced protein levels of p-Erk 1/2 and p-STAT3 in mouse models of pancreatic cancer. All of these proteins are essential for tumor promotion, progression and metastasis. In conclusion, the bromodomain inhibitors JQ1 and I-BET 762 targeted and suppressed multiple pathways in pancreatic cancer. I-BET 762 and a number of other bromodomain inhibitors are currently being tested in several clinical trials, making them potentially promising drugs for the treatment of pancreatic cancer, an often-fatal disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Benzodiazepines/pharmacology , Pancreatic Neoplasms/drug therapy , Triazoles/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Ceruletide , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide/metabolism , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatitis/chemically induced , Pancreatitis/drug therapy , Pancreatitis/immunology , Pancreatitis/metabolism , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects
10.
Carcinogenesis ; 37(12): 1170-1179, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27659181

ABSTRACT

Because the 5-year survival rate for pancreatic cancer remains under 10%, new drugs are needed for the prevention and treatment of this devastating disease. Patients with chronic pancreatitis have a 12-fold higher risk of developing pancreatic cancer. LSL-KrasG12D/+;Pdx-1-Cre (KC) mice replicate the genetics, symptoms and histopathology found in human pancreatic cancer. Immune cells infiltrate into the pancreas of these mice and produce inflammatory cytokines that promote tumor growth. KC mice are particularly sensitive to the effects of lipopolysaccharide (LPS), as only 48% of KC mice survived an LPS challenge while 100% of wildtype (WT) mice survived. LPS also increased the percentage of CD45+ immune cells in the pancreas and immunosuppressive Gr1+ myeloid-derived suppressor cell in the spleen of these mice. The triterpenoid CDDO-imidazolide (CDDO-Im) not only reduced the lethal effects of LPS (71% survival) but also decreased the infiltration of CD45+ cells into the pancreas and the percentage of Gr1+ myeloid-derived suppressor cell in the spleen of KC mice 4-8 weeks after the initial LPS challenge. While the levels of inflammatory cytokine levels were markedly higher in KC mice versus WT mice challenged with LPS, CDDO-Im significantly decreased the production of IL-6, CCL-2, vascular endothelial growth factor and G-CSF in the KC mice. All of these cytokines are prognostic markers in pancreatic cancer or play important roles in the progression of this disease. Disrupting the inflammatory process with drugs such as CDDO-Im might be useful for preventing pancreatic cancer, especially in high-risk populations.


Subject(s)
Inflammation/drug therapy , Neoplasms, Experimental/drug therapy , Pancreas/drug effects , Pancreatic Neoplasms/drug therapy , Animals , Granulocyte Colony-Stimulating Factor/biosynthesis , Homeodomain Proteins/genetics , Humans , Imidazoles/administration & dosage , Inflammation/chemically induced , Inflammation/genetics , Interleukin-6/biosynthesis , Leukocyte Common Antigens/biosynthesis , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Monocyte Chemoattractant Proteins/biosynthesis , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Oleanolic Acid/administration & dosage , Oleanolic Acid/analogs & derivatives , Pancreas/immunology , Pancreas/pathology , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/genetics , Pancreatitis, Chronic/complications , Pancreatitis, Chronic/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Trans-Activators/genetics , Vascular Endothelial Growth Factor A/biosynthesis
11.
Curr Top Med Chem ; 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27320330

ABSTRACT

Rexinoids are selective ligands for the nuclear receptors known as RXRs. They do not bind to the receptors for all-trans-retinoic acid (RARs). Many new rexinoids have been synthesized and then assayed for their ability to suppress proliferation of cancer cells, to inhibit activation of inflammatory cells of the tumor microenvironment, and to prevent carcinogenesis in animal models relevant to human disease. Here we review the literature on the effects of 4 such rexinoids: bexarotene, LG100268, LG101506, and NRX194204. These rexinoids also have potent synergistic effects when used in combination with other active pharmacological agents, and practical clinical applications would benefit from these actions.

13.
Cancer Prev Res (Phila) ; 9(1): 105-14, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26554632

ABSTRACT

LG101506 was originally synthesized to overcome some of the undesirable side effects of rexinoids. We compared the anticarcinogenic action of LG101506 and LG100268 and for the first time showed that both drugs are useful for prevention of lung cancer in A/J mice. These molecules markedly reduced tumor number, tumor size, and total tumor burden, when chronically administered to A/J mice that had been initiated with the mutagenic carcinogen, vinyl carbamate. Moreover, LG100268 synergized with the histone deacetylase inhibitor, vorinostat, for prevention of experimental lung cancer and enhanced the effect of carboplatin/paclitaxel for treatment of experimental lung cancer. Both rexinoids diminished the percentage of high-grade, highly malignant adenocarcinomas found at autopsy. In cell culture studies, the rexinoids exhibited potent anti-inflammatory properties at nanoMolar concentrations. These drugs suppressed the ability of lipopolysaccharide to stimulate the synthesis and secretion of nitric oxide and inflammatory cytokines and chemokines, such as IL6, IL1ß, CXCL2, and CSF3, in macrophage-like RAW264.7 cells. The present results suggest that LG100268, LG101506, or a related rexinoid may have useful clinical applications in the field of oncology.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinogenesis , Fatty Acids, Unsaturated/pharmacology , Lung Neoplasms/drug therapy , Nicotinic Acids/pharmacology , Phenyl Ethers/pharmacology , Tetrahydronaphthalenes/pharmacology , Animals , Carboplatin/administration & dosage , Cell Line , Chemokines/metabolism , Cytokines/metabolism , Female , Flow Cytometry , Humans , Hydroxamic Acids/administration & dosage , Inflammation , Lipopolysaccharides/chemistry , Lipoproteins/blood , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Oligonucleotide Array Sequence Analysis , Paclitaxel/administration & dosage , Random Allocation , Real-Time Polymerase Chain Reaction , U937 Cells , Vorinostat
14.
Pharmacol Res ; 100: 135-47, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26238177

ABSTRACT

Two new analogues of CDDO-Imidazolide (CDDO-Im), namely 1-[2-Cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]-4(-pyridin-2-yl)-1H-imidazole ("CDDO-2P-Im") and 1-[2-Cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]-4(-pyridin-3-yl)-1H-imidazole ("CDDO-3P-Im") have been synthesized and tested for their potential use as chemopreventive drugs. At nanomolar concentrations, they were equipotent to CDDO-Im for inducing differentiation and apoptosis in U937 leukemia cells. As inflammation and oxidative stress contribute to carcinogenesis, we also assessed their cytoprotective potential. The new compounds suppressed inducible nitric oxide synthase (iNOS) expression in RAW264.7 macrophage-like cells and significantly elevated heme oxygenase-1 (HO-1) and quinone reductase (NQO1) mRNA and protein levels in various mouse tissues in vivo. Most importantly, pharmacokinetic studies performed in vitro in human plasma and in vivo showed that each new analogue was more stable than CDDO-Im. Much higher concentrations of the new derivatives were found in mouse liver, lung, pancreas and kidney after gavage in contrast to CDDO-Im. Because of their better bioavailability and their excellent anti-inflammatory profile in vitro, CDDO-2P-Im and CDDO-3P-Im were tested for prevention in a highly relevant mouse lung cancer model, in which A/J mice develop lung carcinomas after injection of vinyl carbamate, a potent carcinogen. CDDO-2P-Im and CDDO-3P-Im were as effective as CDDO-Im for reducing the size and the severity of the lung tumors.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Neoplasms/prevention & control , Oleanolic Acid/analogs & derivatives , Animals , Cell Line, Tumor , Cells, Cultured , Female , Heme Oxygenase-1/metabolism , Humans , Mice , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasms/metabolism , Nitric Oxide Synthase Type II/metabolism , Oleanolic Acid/pharmacology , Oxidative Stress/drug effects , RNA, Messenger/metabolism , U937 Cells
15.
Carcinogenesis ; 36(7): 769-81, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25939751

ABSTRACT

Lung cancer accounts for the highest number of cancer-related deaths in the USA, highlighting the need for better prevention and therapy. Activation of the Nrf2 pathway detoxifies harmful insults and reduces oxidative stress, thus preventing carcinogenesis in various preclinical models. However, constitutive activation of the Nrf2 pathway has been detected in numerous cancers, which confers a survival advantage to tumor cells and a poor prognosis. In our study, we compared the effects of two clinically relevant classes of Nrf2 activators, dimethyl fumarate (DMF) and the synthetic oleanane triterpenoids, CDDO-imidazolide (CDDO-Im) and CDDO-methyl ester (CDDO-Me) in RAW 264.7 mouse macrophage-like cells, in VC1 lung cancer cells and in the A/J model of lung cancer. Although the triterpenoids and DMF both activated the Nrf2 pathway, CDDO-Im and CDDO-Me were markedly more potent than DMF. All of these drugs reduced the production of reactive oxygen species and inhibited nitric oxide production in RAW264.7 cells, but the triterpenoids were 100 times more potent than DMF in these assays. Microarray analysis revealed that only 52 of 99 Nrf2-target genes were induced by all three compounds, and each drug regulated a unique subset of Nrf2 genes. These drugs also altered the expression of other genes important in lung cancer independent of Nrf2. Although all three compounds enhanced the phosphorylation of CREB, only DMF increased the phosphorylation of Akt. CDDO-Me, at either 12.5 or 50mg/kg of diet, was the most effective drug in our lung cancer mouse model. Specifically, CDDO-Me significantly reduced the average tumor number, size and burden compared with the control group (P < 0.05). Additionally, 52% of the tumors in the control group were high-grade tumors compared with only 14% in the CDDO-Me group. Though less potent, CDDO-Im had similar activity as CDDO-Me. In contrast, 61-63% of the tumors in the DMF groups (400-1200mg/kg diet) were high-grade tumors compared with 52% for the controls (P < 0.05). Additionally, DMF significantly increased the average number of tumors compared with the controls (P < 0.05). Thus, in contrast to the triterpenoids, which effectively reduced pathogenesis in A/J mice, DMF enhanced the severity of lung carcinogenesis in these mice. Collectively, these results suggest that although CDDO-Im, CDDO-Me and DMF all activate the Nrf2 pathway, they target distinct genes and signaling pathways, resulting in opposite effects for the prevention of experimental lung cancer.


Subject(s)
Fumarates/pharmacology , Imidazoles/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Oleanolic Acid/analogs & derivatives , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Dimethyl Fumarate , Female , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Inbred Strains , Mice, Knockout , Molecular Targeted Therapy , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neoplasms, Experimental , Oleanolic Acid/pharmacology , Oligonucleotide Array Sequence Analysis , Oxidative Stress/drug effects
16.
J Neurochem ; 133(2): 233-41, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25683606

ABSTRACT

Retinal ischemia plays a critical role in multiple vision-threatening diseases and leads to death of retinal neurons, particularly ganglion cells. Oxidative stress plays an important role in this ganglion cell loss. Nrf2 (NF-E2-related factor 2) is a major regulator of the antioxidant response, and its role in the retina is increasingly appreciated. We investigated the potential retinal neuroprotective function of Nrf2 after ischemia-reperfusion (I/R) injury. In an experimental model of retinal I/R, Nrf2 knockout mice exhibited much greater loss of neuronal cells in the ganglion cell layer than wild-type mice. Primary retinal ganglion cells isolated from Nrf2 knockout mice exhibited decreased cell viability compared to wild-type retinal ganglion cells, demonstrating the cell-intrinsic protective role of Nrf2. The retinal neuronal cell line 661W exhibited reduced cell viability following siRNA-mediated knockdown of Nrf2 under conditions of oxidative stress, and this was associated with exacerbation of increase in reactive oxygen species. The synthetic triterpenoid CDDO-Im (2-Cyano-3,12-dioxooleana-1,9-dien-28-imidazolide), a potent Nrf2 activator, inhibited reactive oxygen species increase in cultured 661W under oxidative stress conditions and increased neuronal cell survival after I/R injury in wild-type, but not Nrf2 knockout mice. Our findings indicate that Nrf2 exhibits a retinal neuroprotective function in I/R and suggest that pharmacologic activation of Nrf2 could be a therapeutic strategy. Oxidative stress is thought to be an important mediator of retinal ganglion cell death in ischemia-reperfusion injury. We found that the transcription factor NF-E2-related factor 2 (Nrf2), a major regulator of oxidative stress, is an important endogenous neuroprotective molecule in retinal ganglion cells in ischemia-reperfusion, exerting a cell-autonomous protective effect.  The triterpenoid 2-Cyano-3,12-dioxooleana-1,9-dien-28-imidazolide (CDDO-Im) reduces neurodegeneration following ischemia-reperfusion in an Nrf2-dependent fashion. This suggests that Nrf2-activating drugs including triterpenoids could be a therapeutic strategy for retinal neuroprotection.


Subject(s)
Ischemia/pathology , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Retinal Ganglion Cells/drug effects , Animals , Cell Survival/genetics , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/genetics , Imidazoles/pharmacology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-E2-Related Factor 2/genetics , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , RNA, Small Interfering/pharmacology , Retina/cytology , Retinal Ganglion Cells/metabolism , tert-Butylhydroperoxide/pharmacology
18.
PLoS One ; 9(9): e107616, 2014.
Article in English | MEDLINE | ID: mdl-25229616

ABSTRACT

Triple-negative breast cancer is associated with poor prognosis because of a high rate of tumor recurrence and metastasis. Previous studies demonstrated that the synthetic triterpenoid, CDDO-Imidazolide (CDDO-Im) induced cell cycle arrest and apoptosis in triple-negative breast cancer. Since a small subpopulation of cancer stem cells has been suggested to be responsible for drug resistance and metastasis of tumors, our present study determined whether the effects of CDDO-Im in triple-negative breast cancer are due to the inhibition of a cancer stem cell subpopulation. CDDO-Im treatment markedly induced cell cycle arrest at G2/M-phase and apoptosis in the triple-negative breast cancer cell lines, SUM159 and MDA-MB-231. Because SUM159 cells were more sensitive to CDDO-Im than MDA-MB-231 cells, the effects of CDDO-Im on the cancer stem cell subpopulation were further investigated in SUM159 cells. SUM159 cells formed tumorspheres in culture, and the cancer stem cell subpopulation, CD24-/EpCAM+ cells, was markedly enriched in SUM159 tumorspheres. The CD24-/EpCAM+ cells in SUM159 tumorspheres were significantly inhibited by CDDO-Im treatment. CDDO-Im also significantly decreased sphere forming efficiency and tumorsphere size in both primary and secondary sphere cultures. PCR array of stem cell signaling genes showed that expression levels of many key molecules in the stem cell signaling pathways, such as Notch, TGF-ß/Smad, Hedgehog and Wnt, were significantly down-regulated by CDDO-Im in SUM159 tumorspheres. Protein levels of Notch receptors (c-Notch1, Notch1 and Notch3), TGF-ß/Smad (pSmad2/3) and Hedgehog downstream effectors (GLI1) also were markedly reduced by CDDO-Im. In conclusion, the present study demonstrates that the synthetic triterpenoid, CDDO-Im, is a potent anti-cancer agent against triple-negative breast cancer cells by targeting the cancer stem cell subpopulation.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Oleanolic Acid/analogs & derivatives , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/pathology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/drug effects , Hedgehog Proteins/metabolism , Humans , M Phase Cell Cycle Checkpoints/drug effects , Oleanolic Acid/pharmacology , Receptors, Notch/metabolism , Transforming Growth Factor beta/metabolism
19.
Org Biomol Chem ; 12(28): 5192-200, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-24915424

ABSTRACT

An efficient synthesis of methyl 2-cyano-3,12-dioxoursol-1,9-dien-28-oate (CDDU-methyl ester) from commercially available ursolic acid, which features an oxidative ozonolysis-mediated C-ring enone formation, and provides the first access to ursolic acid-derived cyano enone analogues with C-ring activation. These new ursolic acid analogues show potent biological activities, with potency of approximately five-fold less than the corresponding oleanolic acid derivatives.


Subject(s)
Anti-Infective Agents/chemical synthesis , Oleanolic Acid/chemistry , Pentacyclic Triterpenes/chemical synthesis , Triterpenes/chemistry , Animals , Anti-Infective Agents/pharmacology , Cell Line , Esters , Inhibitory Concentration 50 , Interferon-gamma/pharmacology , Macrophages/cytology , Macrophages/drug effects , Mice , Molecular Structure , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Oleanolic Acid/pharmacology , Pentacyclic Triterpenes/pharmacology , Triterpenes/pharmacology , Ursolic Acid
20.
J Clin Invest ; 124(6): 2472-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24837432

ABSTRACT

Colitis-associated colon cancer (CAC) develops as a result of inflammation-induced epithelial transformation, which occurs in response to inflammatory cytokine-dependent downregulation of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) and subsequent suppression of prostaglandin metabolism. Agents that both enhance 15-PGDH expression and suppress cyclooxygenase-2 (COX-2) production may more effectively prevent CAC. Synthetic triterpenoids are a class of small molecules that suppress COX-2 as well as inflammatory cytokine signaling. Here, we found that administration of the synthetic triterpenoid 2-cyano-3,12-dioxooleana-1,9(11)-dien-C28-methyl ester (CDDO-Me) suppresses CAC in mice. In a spontaneous, inflammation-driven intestinal neoplasia model, deletion of Smad4 specifically in T cells led to progressive production of inflammatory cytokines, including TNF-α, IFN-γ, iNOS, IL-6, IL-1ß; as well as activation of STAT1 and STAT3; along with suppression of 15-PGDH expression. Oral administration of CDDO-Me to mice with SMAD4-deficient T cells increased survival and suppressed intestinal epithelial neoplasia by decreasing production of inflammatory mediators and increasing expression of 15-PGDH. Induction of 15-PGDH by CDDO-Me was dose dependent in epithelial cells and was abrogated following treatment with TGF-ß signaling inhibitors in vitro. Furthermore, CDDO-Me-dependent 15-PGDH induction was not observed in Smad3-/- mice. Similarly, CDDO-Me suppressed azoxymethane plus dextran sodium sulfate-induced carcinogenesis in wild-type animals, highlighting the potential of small molecules of the triterpenoid family as effective agents for the chemoprevention of CAC in humans.


Subject(s)
Carcinogenesis/drug effects , Carcinogenesis/metabolism , Colonic Neoplasms/prevention & control , Hydroxyprostaglandin Dehydrogenases/biosynthesis , Oleanolic Acid/analogs & derivatives , Animals , Anticarcinogenic Agents/pharmacology , Colitis/complications , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Cyclooxygenase 2/biosynthesis , Enzyme Induction/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/biosynthesis , Oleanolic Acid/pharmacology , STAT Transcription Factors/metabolism , Smad4 Protein/deficiency , Smad4 Protein/genetics , Smad4 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...