Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
J Med Chem ; 64(10): 6996-7032, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33988379

ABSTRACT

We synthesized a directed library of compounds to explore the structure-activity relationships of peroxisome proliferator-activated receptor δ (PPARδ) activation relative to mesenchymal stem cell (MSC) osteogenesis. Our scaffold used para-substituted cinnamic acids as a polar headgroup, a heteroatom and heterocycle core connecting units, and substituted phenyl groups for the lipophilic tail. Compounds were screened for their ability to increase osteogenesis in MSCs, and the most promising were examined for subunit specificity using a quantitative PPAR transactivation assay. Six compounds were selected for in vivo studies in an ovariectomized mouse model of human postmenopausal osteoporosis. Four compounds improved bone density in vivo, with two (12d and 31a) having activity comparable to that of GW0742, a well-studied PPARδ-selective agonist. 31a (2-methyl-4-[N-methyl-N-[5-methylene-4-methyl-2-[4-(trifluoromethyl)phenyl]thiazole]]aminocinnamic acid) had the highest selectivity for PPARδ compared to other subtypes, its selectivity far exceeding that of GW0742. Our results confirm that PPARδ is a new drug target for possible treatment of osteoporosis via in situ manipulation of MSCs.


Subject(s)
Cell Differentiation , Osteogenesis , PPAR delta/agonists , Thiazoles/chemistry , Animals , Cell Differentiation/drug effects , Disease Models, Animal , Drug Design , Female , Femur/diagnostic imaging , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Osteogenesis/drug effects , Osteoporosis/drug therapy , PPAR delta/metabolism , Structure-Activity Relationship , Thiazoles/metabolism , Thiazoles/pharmacology , Thiazoles/therapeutic use , X-Ray Microtomography
2.
Cancer Lett ; 491: 97-107, 2020 10 28.
Article in English | MEDLINE | ID: mdl-32829010

ABSTRACT

Patients with pancreatic adenocarcinoma (PDAC) have a 5-year survival rate of 8%, the lowest of any cancer in the United States. Traditional chemotherapeutic regimens, such as gemcitabine- and fluorouracil-based regimens, often only prolong survival by months. Effective precision targeted therapy is therefore urgently needed to substantially improve survival. In an effort to expedite approval and delivery of targeted therapy to patients, we utilized a platform to develop a novel combination of FDA approved drugs that would target pancreaticoduodenal homeobox1 (PDX1) and baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5) utilizing super-promoters of the target genes to interrogate an FDA approved drug library. We identified and selected metformin, simvastatin and digoxin (C3) as a novel combination of FDA approved drugs, which were shown to effectively target PDX1 and BIRC5 in human PDAC tumors in mice with no toxicity.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Digoxin/administration & dosage , Drug Repositioning , Homeodomain Proteins/antagonists & inhibitors , Metformin/administration & dosage , Pancreatic Neoplasms/drug therapy , Simvastatin/administration & dosage , Survivin/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Drug Combinations , Drug Synergism , High-Throughput Screening Assays , Humans , Male , Mice , Molecular Targeted Therapy , Pancreatic Neoplasms/pathology
3.
J Biol Chem ; 295(29): 9804-9822, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32404366

ABSTRACT

Activation of lipid-burning pathways in the fat-storing white adipose tissue (WAT) is a promising strategy to improve metabolic health and reduce obesity, insulin resistance, and type II diabetes. For unknown reasons, bilirubin levels are negatively associated with obesity and diabetes. Here, using mice and an array of approaches, including MRI to assess body composition, biochemical assays to measure bilirubin and fatty acids, MitoTracker-based mitochondrial analysis, immunofluorescence, and high-throughput coregulator analysis, we show that bilirubin functions as a molecular switch for the nuclear receptor transcription factor peroxisome proliferator-activated receptor α (PPARα). Bilirubin exerted its effects by recruiting and dissociating specific coregulators in WAT, driving the expression of PPARα target genes such as uncoupling protein 1 (Ucp1) and adrenoreceptor ß 3 (Adrb3). We also found that bilirubin is a selective ligand for PPARα and does not affect the activities of the related proteins PPARγ and PPARδ. We further found that diet-induced obese mice with mild hyperbilirubinemia have reduced WAT size and an increased number of mitochondria, associated with a restructuring of PPARα-binding coregulators. We conclude that bilirubin strongly affects organismal body weight by reshaping the PPARα coregulator profile, remodeling WAT to improve metabolic function, and reducing fat accumulation.


Subject(s)
Adipose Tissue, White/metabolism , Bilirubin/pharmacology , Gene Expression Regulation/drug effects , Mitochondria/metabolism , PPAR alpha/metabolism , Animals , Bilirubin/metabolism , Mice , Receptors, Adrenergic, beta-3/biosynthesis , Uncoupling Protein 1/biosynthesis
4.
J Pharm Sci ; 109(9): 2719-2728, 2020 09.
Article in English | MEDLINE | ID: mdl-32473210

ABSTRACT

A new indole based chalcone molecule MOMIPP induced methuosis mediated cell death in gliobastoma and other cancer cell lines. But the drug was insoluble in water and had a very short plasma half-life. The purpose of this work was to develop a formulation that can provide sustained levels of MOMIPP in vivo. Initial studies established drug solubility in various solvents. N-methyl pyrrolidone (NMP) was determined as an excellent solvent for the drug. Subsequently a poloxamer-407 based thermoreversible gel containing NMP was used to develop the formulation. Rheological studies were performed via oscillatory temperature mode, continuous shear analysis, and oscillatory frequency mode experiments. The mechanical properties of the formulations were tested using a texture profile analyzer. The gelation temperature and time of formulations increased with increasing amounts of NMP. However, the viscosity at 20 °C and storage modulus decreased as the amount of NMP increased. Characterization studies helped to identify the gel formulation that was used to administer the drug orally, sub-cutaneously, and intra-peritoneally. When the gel was given intraperitoneally the target plasma and brain levels of over 5 µM was maintained for about 8 h. Thus, a thermoreversible gel formulation that can deliver MOMIPP in animal studies was successfully developed.


Subject(s)
Antineoplastic Agents , Hydrogels , Animals , Brain/metabolism , Gels , Indoles , Poloxamer/metabolism , Pyridines , Rheology , Temperature , Viscosity
5.
Cancer Lett ; 457: 10-19, 2019 08 10.
Article in English | MEDLINE | ID: mdl-31059751

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer mortality with a dismal overall survival rate and an urgent need for detection of minute tumors. Current diagnostic modalities have high sensitivity and specificity for larger tumors, but not for minute PDAC. In this study, we test the feasibility of a precision diagnostic platform for detecting and localizing minute human PDAC in mice. This platform includes: 1) defining BIRC5 as an early PDAC-upregulated gene and utilizing an enhanced BIRC5 super-promoter to drive expression of dual Gaussia luciferase (GLuc) and sr39 thymidine kinase (sr39TK) reporter genes exponentially and specifically in PDAC; 2) utilizing a genetically-engineered AAV2RGD to ensure targeted delivery of GLuc and sr39TK specifically to PDAC; 3) using serologic GLuc and sr39TK microPET/CT imaging to detect and localize minute human PDAC in mice. The study demonstrates feasibility of a precision diagnostic platform using an integrated technology through a multiple-stage amplification strategy of dual reporter genes to enhance the specificity and sensitivity of detection and localization of minute PDAC tumors and currently undetectable disease.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/diagnostic imaging , Molecular Imaging , Optical Imaging , Pancreatic Neoplasms/diagnostic imaging , Positron Emission Tomography Computed Tomography , Survivin/metabolism , X-Ray Microtomography , Animals , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Feasibility Studies , Gene Expression Regulation, Neoplastic , Humans , Luciferases/genetics , Luciferases/metabolism , Male , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Predictive Value of Tests , Promoter Regions, Genetic , Survivin/genetics , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Tumor Burden , Up-Regulation
6.
BMC Cancer ; 19(1): 77, 2019 Jan 16.
Article in English | MEDLINE | ID: mdl-30651087

ABSTRACT

BACKGROUND: Synthetic indolyl- pyridinyl- propenones (IPPs) induce methuosis, a form of non-apoptotic cell death, in glioblastoma and other cancer cell lines. Methuosis is characterized by accumulation of cytoplasmic vacuoles derived from macropinosomes and late endosomes, followed by metabolic failure and rupture of the plasma membrane. However, not all IPPs that cause vacuolization are cytotoxic. The main goals of the present study were to identify key signaling pathways that contribute to methuosis induced by cytotoxic IPPs and to evaluate the anti-tumor potential of a prototype IPP in vivo. METHODS: We utilized metabolic flux analysis, glucose uptake, immunoblotting, and selective pharmacological inhibitors to compare the effects of closely related cytotoxic and non-cytotoxic IPPs in cultured glioblastoma cells. To determine whether the use of methuosis-inducing IPPs might be feasible in a therapeutic context, we quantified the distribution of our lead IPP compound, MOMIPP, in mouse plasma and brain, and tested its ability to inhibit tumor growth in an intracerebral glioblastoma xenograft model. RESULTS: The cytotoxic IPP compound, MOMIPP, causes early disruptions of glucose uptake and glycolytic metabolism. Coincident with these metabolic changes, MOMIPP selectively activates the JNK1/2 stress kinase pathway, resulting in phosphorylation of c-Jun, Bcl-2 and Bcl-xL. At the same concentration, the non-cytotoxic analog, MOPIPP, does not activate these pathways. Pharmacologic inhibition of JNK activity promotes survival, even when cells are extensively vacuolated, but suppression of c-Jun transcriptional activity offers no protection. MOMIPP readily penetrates the blood-brain barrier and is moderately effective in suppressing progression of intracerebral glioblastoma xenografts. CONCLUSIONS: The results suggest that interference with glucose uptake and induction of JNK-mediated phosphorylation of pro-survival members of the Bcl-2 family represent key events in the methuosis death process. In addition to providing new insights into the underlying molecular mechanism of methuosis, the results indicate that compounds of the cytotoxic IPP class may have potential for further development as therapeutic agents for brain tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death/drug effects , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Pyridines/pharmacology , Adult , Animals , Antineoplastic Agents/therapeutic use , Brain/metabolism , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Indoles/therapeutic use , Mice , Pyridines/therapeutic use , Tissue Distribution , Xenograft Model Antitumor Assays
7.
Cancer Chemother Pharmacol ; 83(2): 237-254, 2019 02.
Article in English | MEDLINE | ID: mdl-30426158

ABSTRACT

PURPOSE: 3-(6-Methoxy-2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propene-1-one (6-MOMIPP) is a novel indole-based chalcone that disrupts microtubules. The present study aims to define the mechanism through which 6-MOMIPP induces cell death and to evaluate the efficacy of the compound in penetrating the blood-brain barrier and inhibiting growth of glioblastoma xenografts. METHODS: The effects of 6-MOMIPP were evaluated in cultured U251 glioblastoma cells, using viability, flow cytometry, and tubulin polymerization assays. Scintillation proximity and tubulin crosslinking methods were used to identify the binding site of 6-MOMIPP on tubulin, and western blots were performed to define the signaling pathways that contribute to cell death. LC/MS assays were used to study the pharmacokinetic behavior of 6-MOMIPP in mice. Subcutaneous and intracerebral xenograft models were utilized to assess the effects of 6-MOMIPP on growth of U251 glioblastoma in vivo. RESULTS: The findings indicate that 6-MOMIPP targets the colchicine site on ß-tubulin. At concentrations ≥ 250 nm, 6-MOMIPP induces mitotic arrest, caspase activation and loss of cell viability. Cells are protected by caspase inhibitors, pointing to an apoptotic mechanism of cell death. Loss of cell viability is preceded by activation of Cdk1(Cdc2) and phosphorylation of Bcl-2 and Bcl-xL. Inhibition of both events with a Cdk1 inhibitor prevents cell death. 6-MOMIPP has broad activity against the viability of multiple glioblastoma, melanoma and lung carcinoma cell lines. Viability of normal cells, including differentiated neurons, is not significantly affected at a drug concentration (1 µM) that reduces viability in most cancer lines. Pharmacokinetic studies in mice show that concentrations of 6-MOMIPP in the brain mirror those in the plasma, indicating that 6-MOMIPP readily penetrates the blood-brain barrier. Studies with mice bearing human U251 glioblastoma xenografts demonstrate that 6-MOMIPP is effective in suppressing growth of subcutaneous and intracerebral tumors without causing general toxicity. CONCLUSIONS: The results indicate that 6-MOMIPP is a novel microtubule disruptor that targets the colchicine binding site on ß-tubulin to induce mitotic arrest and cell death. The ability of 6-MOMIPP to penetrate the blood-brain barrier and inhibit growth of glioblastoma xenografts suggests that it warrants further preclinical evaluation as potential small-molecule therapeutic that may have advantages in treating primary and metastatic brain tumors.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/drug therapy , Cell Proliferation/drug effects , Glioblastoma/drug therapy , Indoles/pharmacology , Mitosis , Pyridines/pharmacology , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle/drug effects , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Indoles/pharmacokinetics , Mice , Mice, Nude , Pyridines/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Nutrients ; 10(11)2018 Nov 03.
Article in English | MEDLINE | ID: mdl-30400270

ABSTRACT

The 5'-3' structure-specific endonuclease ERCC1/XPF (Excision Repair Cross-Complementation Group 1/Xeroderma Pigmentosum group F) plays critical roles in the repair of cisplatin-induced DNA damage. As such, it has been identified as a potential pharmacological target for enhancing clinical response to platinum-based chemotherapy. The goal of this study was to follow up on our previous identification of the compound NSC143099 as a potent inhibitor of ERCC1/XPF activity by performing an in silico screen to identify structural analogues that could inhibit ERCC1/XPF activity in vitro and in vivo. Using a fluorescence-based DNA-endonuclease incision assay, we identified the green tea polyphenol (-)-epigallocatechin-3-gallate (EGCG) as a potent inhibitor of ERCC1/XPF activity with an IC50 (half maximal inhibitory concentration) in the nanomolar range in biochemical assays. Using DNA repair assays and clonogenic survival assays, we show that EGCG can inhibit DNA repair and enhance cisplatin sensitivity in human cancer cells. Finally, we show that a prodrug of EGCG, Pro-EGCG (EGCG octaacetate), can enhance response to platinum-based chemotherapy in vivo. Together these data support a novel target of EGCG in cancer cells, namely ERCC1/XPF. Our studies also corroborate previous observations that EGCG enhances sensitivity to cisplatin in multiple cancer types. Thus, EGCG or its prodrug makes an ideal candidate for further pharmacological development with the goal of enhancing cisplatin response in human tumors.


Subject(s)
Catechin/analogs & derivatives , Cisplatin/pharmacology , DNA Repair/drug effects , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Polyphenols/pharmacology , Animals , Apoptosis/drug effects , Catechin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Comet Assay , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm , Endonucleases/genetics , Female , Humans , Mice , Mice, Nude , Platinum/pharmacology , Prodrugs/pharmacology , Tea/chemistry
10.
Nat Commun ; 8: 15559, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28555620

ABSTRACT

Inflammation and thrombosis occur together in many diseases. The leukocyte integrin Mac-1 (also known as integrin αMß2, or CD11b/CD18) is crucial for leukocyte recruitment to the endothelium, and Mac-1 engagement of platelet GPIbα is required for injury responses in diverse disease models. However, the role of Mac-1 in thrombosis is undefined. Here we report that mice with Mac-1 deficiency (Mac-1-/-) or mutation of the Mac-1-binding site for GPIbα have delayed thrombosis after carotid artery and cremaster microvascular injury without affecting parameters of haemostasis. Adoptive wild-type leukocyte transfer rescues the thrombosis defect in Mac-1-/- mice, and Mac-1-dependent regulation of the transcription factor Foxp1 contributes to thrombosis as evidenced by delayed thrombosis in mice with monocyte-/macrophage-specific overexpression of Foxp1. Antibody and small-molecule targeting of Mac-1:GPIbα inhibits thrombosis. Our data identify a new pathway of thrombosis involving leukocyte Mac-1 and platelet GPIbα, and suggest that targeting this interaction has anti-thrombotic therapeutic potential with reduced bleeding risk.


Subject(s)
Blood Platelets/immunology , Leukocytes/metabolism , Macrophage-1 Antigen/genetics , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombosis/immunology , Animals , Binding Sites , Bleeding Time , Blood Coagulation , Blood Platelets/cytology , Blood Platelets/metabolism , Carotid Arteries/pathology , Glucosamine/chemistry , Hemostasis , Inflammation , Leukocytes/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microcirculation , NIH 3T3 Cells , Partial Thromboplastin Time , Phagocytosis , Platelet Activation , Platelet Count , Protein Binding , Protein Domains , Signal Transduction , Thrombin/metabolism
11.
Eur J Med Chem ; 122: 79-91, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27343855

ABSTRACT

Certain indolyl-pyridinyl-propenone analogues kill glioblastoma cells that have become resistant to conventional therapeutic drugs. Some of these analogues induce a novel form of non-apoptotic cell death called methuosis, while others primarily cause microtubule disruption. Ready access to 5-indole substitution has allowed characterization of this position to be important for both types of mechanisms when a simple methoxy group is present. We now report the syntheses and biological effects of isomeric methoxy substitutions on the indole ring. Additionally, analogues containing a trimethoxyphenyl group in place of the pyridinyl moiety were evaluated for anticancer activity. The results demonstrate that the location of the methoxy group can alter both the potency and the mechanism of cell death. Remarkably, changing the methoxy from the 5-position to the 6-position switched the biological activity from induction of methuosis to disruption of microtubules. The latter may represent a prototype for a new class of mitotic inhibitors with potential therapeutic utility.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Indoles/chemical synthesis , Indoles/pharmacology , Pyridines/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Humans , Indoles/chemistry , Isomerism , Structure-Activity Relationship
12.
J Nat Prod ; 78(12): 2940-7, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26654660

ABSTRACT

Stressed soybeans produce a group of phytoalexins that belong to the 6a-hydroxypterocarpan family of flavonoids. Certain of the more prominent members, such as the glyceollins I, II, and III, have demonstrated potential antidiabetic properties and promising cytotoxicity in both human breast and prostate cancer cell cultures with preliminary studies in animals further demonstrating antitumor effects in estrogen-dependent, human breast cancer cell implants. Although syntheses of glyceollin I have been reported previously, this work constitutes the first total directed synthesis of (±)-glyceollin II. It involves 12 steps with an overall yield of 7% using practical methods that should be readily scalable to produce quantities needed for advanced biological characterization. Highlights include a novel intramolecular benzoin condensation, a chelation-controlled lithium aluminum hydride-mediated reduction, and an intramolecular cyclization via the formation of a transient epoxide intermediate to cap the construction of the 6a-hydroxypterocarpan system. Additionally, a dihydro analogue has been obtained, and several isolated intermediates have been made available for evaluation of their biological properties and possible contributions toward elaborating key structure-activity relationship data among this family of promising phytoalexins elicited from stressed soybeans.


Subject(s)
Antineoplastic Agents, Phytogenic/chemical synthesis , Pterocarpans/chemical synthesis , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Benzopyrans , Breast Neoplasms/drug therapy , Estrogens/pharmacology , Female , Heterocyclic Compounds, 4 or More Rings , Humans , Male , Molecular Structure , Prostatic Neoplasms/drug therapy , Pterocarpans/chemistry , Pterocarpans/pharmacology , Sesquiterpenes , Glycine max/chemistry , Stereoisomerism , Structure-Activity Relationship , Phytoalexins
13.
J Med Chem ; 58(5): 2489-512, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25654321

ABSTRACT

Methuosis is a form of nonapoptotic cell death characterized by an accumulation of macropinosome-derived vacuoles with eventual loss of membrane integrity. Small molecules inducing methuosis could offer significant advantages compared to more traditional anticancer drug therapies that typically rely on apoptosis. Herein we further define the effects of chemical substitutions at the 2- and 5-indolyl positions on our lead compound 3-(5-methoxy-2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propene-1-one (MOMIPP). We have identified a number of compounds that induce methuosis at similar potencies, including an interesting analogue having a hydroxypropyl substituent at the 2-position. In addition, we have discovered that certain substitutions on the 2-indolyl position redirect the mode of cytotoxicity from methuosis to microtubule disruption. This switch in activity is associated with an increase in potency as large as 2 orders of magnitude. These compounds appear to represent a new class of potent microtubule-active anticancer agents.


Subject(s)
Alkenes/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Glioblastoma/drug therapy , Indoles/chemistry , Microtubules/drug effects , Pyridines/chemistry , Cell Cycle/drug effects , Cell Proliferation/drug effects , Glioblastoma/pathology , Humans , Indoles/pharmacology , Microscopy, Fluorescence , Pyridines/pharmacology , Structure-Activity Relationship , Tubulin/metabolism , Tumor Cells, Cultured
14.
ACS Med Chem Lett ; 5(1): 73-77, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24527179

ABSTRACT

Because many cancers harbor mutations that confer resistance to apoptosis, there is a need for therapeutic agents that can trigger alternative forms of cell death. Methuosis is a novel form of non-apoptotic cell death characterized by accumulation of vacuoles derived from macropinosomes and endosomes. Previous studies identified an indole-based chalcone, 3-(5-methoxy-2-methylindol-3-yl)-1-(4-pyridinyl)-2-propen-1-one (MOMIPP), that induces methuosis in human cancer cells. Herein, we describe the synthesis of related 2-indolyl substituted pyridinylpropenones and their effects on U251 glioblastoma cells. Increasing the size of the 2-indolyl substituent substantially reduces growth inhibitory activity and cytotoxicity, but does not prevent cell vacuolization. Computational models suggest that the results are not due to steric-driven conformational effects. The unexpected uncoupling of vacuolization and cell death implies that the relationship between endosomal perturbations and methuotic cell death is more complex than previously realized. The new series of compounds will be useful in further defining the molecular and cellular mechanisms underlying methuosis.

15.
Eur J Med Chem ; 63: 104-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23474897

ABSTRACT

Retinoic acid receptor alpha (RARα) selective compounds may guide the design of drugs that can be used in conjunction with hormonal adjuvant therapy in the treatment of breast cancer. Herein we report a modified synthesis of a known RARα antagonist, 2-fluoro-4-[[[8-bromo-2,2-dimethyl-4-(4-methylphenyl)chroman-6-yl]carbonyl]amino]benzoic acid and a synthesis of its unknown, desfluoro analog, 4-[[[8-bromo-2,2-dimethyl-4-(4-methylphenyl)chroman-6-yl]carbonyl]amino]benzoic acid. The modified route allows for facile reaction workups, increased yields, lower cost and incorporates a green alternative step. Structure-activity relationship studies determined through functional cell-based assays, demonstrated antagonism to RARα for both compounds. Molecular modeling within the RARα binding pocket was used to compare binding interactions of the desfluoro analog to a known RAR antagonist.


Subject(s)
Chromans/chemical synthesis , Chromans/pharmacology , Receptors, Retinoic Acid/antagonists & inhibitors , Benzoates/chemical synthesis , Benzoates/chemistry , Benzoates/pharmacology , Binding Sites , Chromans/chemistry , Dose-Response Relationship, Drug , Humans , MCF-7 Cells , Models, Chemical , Models, Molecular , Molecular Structure , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Structure-Activity Relationship , Transcriptional Activation/drug effects , Tretinoin/pharmacology
16.
J Agric Food Chem ; 60(25): 6376-82, 2012 Jun 27.
Article in English | MEDLINE | ID: mdl-22655912

ABSTRACT

Soy glyceollins, induced during stress, have been shown to inhibit cancer cell growth in vitro and in vivo. In the present study, we used prediabetic rats to examine the glyceollins effect on blood glucose. During an oral glucose tolerance test (OGTT), the blood glucose excursion was significantly decreased in the rats treated with oral administration of either 30 or 90 mg/kg glyceollins. Plasma analysis demonstrated that glyceollins are absorbed after oral administration, and duration of exposure extends from 20 min to at least 4 h postadministration. Exposure of 3T3-L1 adipocytes to glyceollins significantly increased both insulin-stimulated and basal glucose uptake. Basal glucose uptake was increased 1.5-fold by exposure to 5 µM glyceollin in a dose-response manner. Coincubation with insulin significantly stimulated maximal glucose uptake above basal uptake levels and tended to increase glucose uptake beyond the levels of either stimulus alone. On a molecular level, polymerase chain reaction showed significantly increased levels of glucose transporter GLUT4 mRNA in 3T3-L1 adipocytes, especially when the cells were exposed to 5 µM glyceollins for 3 h in vitro. It correlated with elevated protein levels of GLUT4 detected in the 5 µM glyceollin-treated cells. Thus, the simulative effect of the glyceollins on adipocyte glucose uptake was attributed to up-regulation of glucose transporters. These findings indicate potential benefits of the glyceollins as an intervention in prediabetic conditions as well as a treatment for type 1 and type 2 diabetes by increasing both the insulin-mediated and the basal, insulin-independent, glucose uptake by adipocytes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucose/metabolism , Glycine max/chemistry , Isoflavones/administration & dosage , Plant Extracts/administration & dosage , Pterocarpans/administration & dosage , Sesquiterpenes/administration & dosage , 3T3 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Biological Transport , Diabetes Mellitus, Type 2/metabolism , Humans , Male , Mice , Rats , Rats, Sprague-Dawley , Phytoalexins
17.
J Phys Chem B ; 116(18): 5425-36, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22510106

ABSTRACT

Association energies of the acetate ion with cationic amines bearing one to three methyl groups were calculated in the range of -14 to -17 kcal/mol in aqueous solution by means of the IEF-PCM method at the CCSD(T)/CBS//MP2/aug-cc-pvdz and DFT/B97D/CBS//B97D/aug-cc-pvtz levels. The main stabilization factor for the association is the possibility for the formation of an ionic intermolecular hydrogen bond between the elements of the complex. For a quaternary ammonium ion, the favorable electrostatic interaction energy is the only driving force, and the stabilization energy for the complex is reduced to -4 kcal/mol. The internal free energies of the ion-pair tautomers of the studied species are higher by 10-15 kcal/mol in water than those for the neutral, hydrogen-bonded forms. Monte Carlo free energy perturbation calculations at T = 298 K and p = 1 atm predict -11 to -16 kcal/mol relative solvation free energy in favor of the corresponding ionic form. As a result, the ion-pair tautomer is the prevailing form in aqueous solution and on the extracellular surface of a receptor. Modeling the complex of a protonated ligand interacting with an Asp/Glu carboxylate side-chain in the binding cavity of a receptor, two strongly bound water molecules were considered so as to form hydrogen-bonded water bridges between the elements of the ion-pair. Nonetheless, the low polarity environment mimicked by a chloroform solvent cannot stabilize the ionic tautomer. A proton jump was predicted, which suggests that acetylcholine, an inherent cation by structure, might have evolved as the natural agonist for muscarinic receptors because a quaternary ammonium system assures the maintenance of the ion-pair form with a carboxylate side-chain in a protein cavity, the latter perhaps then being needed for receptor activation.


Subject(s)
Amines/chemistry , Carboxylic Acids/chemistry , Quaternary Ammonium Compounds/chemistry , Acetates/chemistry , Hydrogen Bonding , Monte Carlo Method , Solutions , Solvents/chemistry , Surface Properties , Thermodynamics , Water/chemistry
18.
Bioorg Med Chem ; 20(9): 2950-6, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22464683

ABSTRACT

Microbes that have gained resistance against antibiotics pose a major emerging threat to human health. New targets must be identified that will guide the development of new classes of antibiotics. The selective inhibition of key microbial enzymes that are responsible for the biosynthesis of essential metabolites can be an effective way to counter this growing threat. Aspartate semialdehyde dehydrogenases (ASADHs) produce an early branch point metabolite in a microbial biosynthetic pathway for essential amino acids and for quorum sensing molecules. In this study, molecular modeling and docking studies were performed to achieve two key objectives that are important for the identification of new selective inhibitors of ASADH. First, virtual screening of a small library of compounds was used to identify new core structures that could serve as potential inhibitors of the ASADHs. Compounds have been identified from diverse chemical classes that are predicted to bind to ASADH with high affinity. Next, molecular docking studies were used to prioritize analogs within each class for synthesis and testing against representative bacterial forms of ASADH from Streptococcus pneumoniae and Vibrio cholerae. These studies have led to new micromolar inhibitors of ASADH, demonstrating the utility of this molecular modeling and docking approach for the identification of new classes of potential enzyme inhibitors.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Aspartate-Semialdehyde Dehydrogenase/metabolism , Binding Sites , Enzyme Inhibitors/chemical synthesis , Kinetics , Molecular Dynamics Simulation , Protein Structure, Tertiary , Streptococcus pneumoniae/enzymology , Vibrio cholerae/enzymology
19.
J Biomol Screen ; 17(5): 673-82, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22460173

ABSTRACT

The rise in organisms resistant to existing drugs has added urgency to the search for new antimicrobial agents. Aspartate ß-semialdehyde dehydrogenase (ASADH) catalyzes a critical step in an essential microbial pathway that is absent in mammals. Our laboratory is using fragment library screening to identify efficient and selective ASADH inhibitors. These preliminary agents are then tested to identify compounds with desired antimicrobial properties for further refinement. Toward this end, we have established a microplate-based, dual-assay approach using a single reagent to evaluate antibiotic activity and mammalian cell toxicity during early stage development. The bacterial assay uses nonpathogenic bacteria to allow efficacy testing without a dedicated microbial laboratory. Toxicity assays are performed with a panel of mammalian cells derived from representative susceptible tissues. These assays can be adapted to target other microbial systems, such as fungi and biofilms, and additional mammalian cell lines can be added as needed. Application of this screening approach to antibiotic standards demonstrates the ability of these assays to identify bacterial selectivity and potential toxicity issues. Tests with selected agents from the ASADH inhibitor fragment library show some compounds with antibiotic activity, but as expected, most of these early agents display higher than desired mammalian cell toxicity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Microbial Sensitivity Tests/methods , Anti-Bacterial Agents/toxicity , Cell Line , Enzyme Inhibitors/toxicity , Humans , Inhibitory Concentration 50 , Reproducibility of Results
20.
J Med Chem ; 55(5): 1940-56, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22335538

ABSTRACT

Methuosis is a novel caspase-independent form of cell death in which massive accumulation of vacuoles derived from macropinosomes ultimately causes cells to detach from the substratum and rupture. We recently described a chalcone-like compound, 3-(2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propen-1-one (i.e., MIPP), which can induce methuosis in glioblastoma and other types of cancer cells. Herein, we describe the synthesis and structure-activity relationships of a directed library of related compounds, providing insights into the contributions of the two aryl ring systems and highlighting a potent derivative, 3-(5-methoxy, 2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propen-1-one (i.e., MOMIPP) that can induce methuosis at low micromolar concentrations. We have also generated biologically active azide derivatives that may be useful for future studies aimed at identifying the protein targets of MOMIPP by photoaffinity labeling techniques. The potential significance of these studies is underscored by the finding that MOMIPP effectively reduces the growth and viability of Temozolomide-resistant glioblastoma and doxorubicin-resistant breast cancer cells. Thus, it may serve as a prototype for drugs that could be used to trigger death by methuosis in cancers that are resistant to conventional forms of cell death (e.g., apoptosis).


Subject(s)
Antineoplastic Agents/chemical synthesis , Azides/chemical synthesis , Chalcones/chemical synthesis , Indoles/chemical synthesis , Pyridines/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Azides/chemistry , Azides/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chalcones/chemistry , Chalcones/pharmacology , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Indoles/chemistry , Indoles/pharmacology , Photoaffinity Labels/chemical synthesis , Photoaffinity Labels/chemistry , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...