Network


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

Hotspot


Dive into the research topics where Dale G. Nagle is active.

Publication


Featured researches published by Dale G. Nagle.


Bioorganic & Medicinal Chemistry | 2010

The marine sponge metabolite mycothiazole: A novel prototype mitochondrial complex I inhibitor

J. Brian Morgan; Fakhri Mahdi; Yang Liu; Veena Coothankandaswamy; Mika B. Jekabsons; Tyler A. Johnson; Koneni V. Sashidhara; Phillip Crews; Dale G. Nagle; Yu-Dong Zhou

A natural product chemistry-based approach was applied to discover small-molecule inhibitors of hypoxia-inducible factor-1 (HIF-1). A Petrosaspongia mycofijiensis marine sponge extract yielded mycothiazole (1), a solid tumor selective compound with no known mechanism for its cell line-dependent cytotoxic activity. Compound 1 inhibited hypoxic HIF-1 signaling in tumor cells (IC(50) 1nM) that correlated with the suppression of hypoxia-stimulated tumor angiogenesis in vitro. However, 1 exhibited pronounced neurotoxicity in vitro. Mechanistic studies revealed that 1 selectively suppresses mitochondrial respiration at complex I (NADH-ubiquinone oxidoreductase). Unlike rotenone, MPP(+), annonaceous acetogenins, piericidin A, and other complex I inhibitors, mycothiazole is a mixed polyketide/peptide-derived compound with a central thiazole moiety. The exquisite potency and structural novelty of 1 suggest that it may serve as a valuable molecular probe for mitochondrial biology and HIF-mediated hypoxic signaling.


Current Medicinal Chemistry | 2004

Mechanism Targeted Discovery of Antitumor Marine Natural Products

Dale G. Nagle; Yu-Dong Zhou; Flor D. Mora; Kaleem A. Mohammed; Yong-Pil Kim

Antitumor drug discovery programs aim to identify chemical entities for use in the treatment of cancer. Many strategies have been used to achieve this objective. Natural products have always played a major role in anticancer medicine and the unique metabolites produced by marine organisms have increasingly become major players in antitumor drug discovery. Rapid advances have occurred in the understanding of tumor biology and molecular medicine. New insights into mechanisms responsible for neoplastic disease are significantly changing the general philosophical approach towards cancer treatment. Recently identified molecular targets have created exciting new means for disrupting tumor-specific cell signaling, cell division, energy metabolism, gene expression, drug resistance and blood supply. Such tumor-specific treatments could someday decrease our reliance on traditional cytotoxicity-based chemotherapy and provide new less toxic treatment options with significantly fewer side effects. Novel molecular targets and state-of-the-art, molecular mechanism-based screening methods have revitalized antitumor research and these changes are becoming an ever-increasing component of modern antitumor marine natural products research. This review describes marine natural products identified using tumor-specific mechanism-based assays for regulators of angiogenesis, apoptosis, cell cycle, macromolecule synthesis, mitochondrial respiration, mitosis, multidrug efflux and signal transduction. Special emphasis is placed on natural products directly discovered using molecular mechanism-based screening.


Tetrahedron Letters | 1995

Absolute configuration of curacin A, a novel antimitotic agent from the tropical marine cyanobacterium Lyngbya majuscula

Dale G. Nagle; Robin S. Geralds; Hye-Dong Yoo; William H. Gerwick; Tae-Seong Kim; Mitch Nambu; James D. White

Curacin A is a structurally novel antimitotic agent isolated from the Caribbean cyanobacterium Lyngbya majuscula. Its planar structure has been previously determined from a spectroscopic investigation. Here, we define the complete relative and absolute configuration of curacin A by comparison of products obtained from chemical degradation of the natural product with the same substances prepared by synthesis. Curacin A is shown to have 2R, 13R, 19R, 21S absolute configuration.


Tetrahedron Letters | 1990

Isolation and structure of constanolactones A and B, new cyclopropyl hydroxy-eicosanoids from the temperate red alga constantinea simplex

Dale G. Nagle; William H. Gerwick

Two new cyclopropyl and lactone containing eicosanoids, constanolactone A and B, were isolated as synthetic diacetate derivatives from the temperate red alga Constantinea simplex and their structures determined by spectroscopic means. A hydrolysis product of one of these helped to define the lactone size in these new eicosanoids. The co-occurrence of three known 12-lipoxygenase metabolites, 12-S-HETE, 12-S-HEPE, and 12-oxododeca-5(Z), 8(E), 10(E)-trienoic acid, supports a 12-1ipoxygenase origin for the new compounds.


Journal of Natural Products | 2008

Latrunculin A and Its C-17-O-Carbamates Inhibit Prostate Tumor Cell Invasion and HIF-1 Activation in Breast Tumor Cells

Khalid A. El Sayed; Mohammad A. Khanfar; Hassan M. Shallal; Anbalagan Muralidharan; Bhushan Awate; Diaa T. A. Youssef; Yang Liu; Yu-Dong Zhou; Dale G. Nagle; Girish V. Shah

The marine-derived macrolides latrunculins A ( 1) and B, from the Red Sea sponge Negombata magnifica, have been found to reversibly bind actin monomers, forming a 1:1 complex with G-actin and disrupting its polymerization. The microfilament protein actin is responsible for several essential functions within the cell such as cytokinesis and cell migration. One of the main binding pharmacophores of 1 to G-actin was identified as the C-17 lactol hydroxyl moiety that binds arginine 210 NH. Latrunculin A-17- O-carbamates 2- 6 were prepared by reaction with the corresponding isocyanates. Latrunculin A ( 1) and carbamates 4- 6 displayed potent anti-invasive activity against the human highly metastatic human prostate cancer PC-3M cells in a Matrigel assay at a concentration range of 50 nM to 1 microM. Latrunculin A ( 1, 500 nM) decreased the disaggregation and cell migration of PC-3M-CT+ spheroids by 3-fold. Carbamates 4 and 5 were 2.5- and 5-fold more active than 1, respectively, in this assay with less actin binding affinity. Latrunculin A ( 1, IC 50 6.7 microM) and its 17- O-[ N-(benzyl)carbamate ( 6, IC 50 29 microM) suppress hypoxia-induced HIF-1 activation in T47D breast tumor cells.


Journal of Biological Chemistry | 2009

Methylalpinumisoflavone Inhibits Hypoxia-inducible Factor-1 (HIF-1) Activation by Simultaneously Targeting Multiple Pathways

Yang Liu; Coothan K. Veena; J. Brian Morgan; Kaleem A. Mohammed; Mika B. Jekabsons; Dale G. Nagle; Yu-Dong Zhou

Hypoxia is a common feature of solid tumors, and the extent of tumor hypoxia correlates with advanced disease stages and treatment resistance. The transcription factor hypoxia-inducible factor-1 (HIF-1) represents an important tumor-selective molecular target for anticancer drug discovery directed at tumor hypoxia. A natural product chemistry-based approach was employed to discover small molecule inhibitors of HIF-1. Bioassay-guided isolation of an active lipid extract of the tropical legumaceous plant Lonchocarpus glabrescens and structure elucidation afforded two new HIF-1 inhibitors: alpinumisoflavone (compound 1) and 4′-O-methylalpinumisoflavone (compound 2). In human breast tumor T47D cells, compounds 1 and 2 inhibited hypoxia-induced HIF-1 activation with IC50 values of 5 and 0.6 μm, respectively. At the concentrations that in hibited HIF-1 activation, compound 2 inhibited hypoxic induction of HIF-1 target genes (CDKN1A, GLUT-1, and VEGF), tumor angiogenesis in vitro, cell migration, and chemotaxis. Compound 2 inhibits HIF-1 activation by blocking the induction of nuclear HIF-1α protein, the oxygen-regulated subunit that controls HIF-1 activity. Mechanistic studies indicate that, unlike rotenone and other mitochondrial inhibitors, compound 2 represents the first small molecule that inhibits HIF-1 activation by simultaneously suppressing mitochondrial respiration and disrupting protein translation in vitro. This unique mechanism distinguishes compound 2 from other small molecule HIF-1 inhibitors that are simple mitochondrial inhibitors or flavanoid-based protein kinase inhibitors.


Hydrobiologia | 1993

Biologically active oxylipins from seaweeds

William H. Gerwick; Philip J. Proteau; Dale G. Nagle; Mitchell L. Wise; Zhi D. Jiang; Matthew W. Bernart; Mats Hamberg

Our previous research has shown that many red algae metabolize polyunsaturated fatty acids to oxidized products resembling the eicosanoid hormones from mammals. We have extended these studies to members of the Phaeophyceae and Chlorophyta and find they also possess similar biosynthetic pathways. From several we have identified novel prostaglandin-like substances. Studies of the molecular mechanisms by which some of these marine oxylipins are formed have revealed that novel oxidative reactions are utilized. Understanding of these biosynthetic pathways in detail has allowed their utilization to produce research biochemicals of high value, such as 12S-hydroperoxyeicosatetraenoic acid (12S-HPETE). Because of their biological properties, seaweed-derived oxylipins have potential utility as pharmaceuticals and research biochemicals.


Phytochemistry Reviews | 2009

Marine natural products as inhibitors of hypoxic signaling in tumors

Dale G. Nagle; Yu-Dong Zhou

Marine natural products have become a major source of new chemical entities in the discovery of potential anticancer agents that potently suppress various antitumor molecular targets. As a consequence of insufficient vascularization, hypoxic regions form within rapidly growing solid tumor masses. Specific alterations of gene expression in these hypoxic tumor cells help facilitate the survival and metastatic spread of solid tumors. The transcriptional response to cellular hypoxia is primarily mediated by the transcription factor hypoxia-inducible factor-1 (HIF-1) that regulates the expression of more than 100 genes involved in cellular adaptation and survival under hypoxic stress. Clinical studies in cancer patients indicate that HIF-1 activation is directly correlated with advanced disease stages and treatment resistance. HIF-1 has emerged as an important tumor-selective molecular target for anticancer drug discovery. As a result, natural product-based inhibitors of HIF-1 activation have been identified from plants and microorganisms. Recently, structurally unique natural products from marine sponges, crinoids, and algae have been identified as HIF-1 activation inhibitors. The US National Cancer Institute’s Open Repository of marine invertebrate and algae extracts has proven to be a valuable source of natural product HIF-1 inhibitors. Among the active compounds identified, certain marine natural products have also been shown to suppress the hypoxic induction of HIF-1 target genes such as vascular endothelial growth factor (VEGF). Some of these marine HIF-1 inhibitors act by interfering with the generation of mitochondrial signaling molecules in hypoxic cells. However, the precise mechanisms of action for many newly identified marine natural product HIF-1 inhibitors remain unresolved.


Journal of Natural Products | 2009

The Caulerpa pigment caulerpin inhibits HIF-1 activation and mitochondrial respiration.

Yang Liu; J. Brian Morgan; Veena Coothankandaswamy; Rui Liu; Mika B. Jekabsons; Fakhri Mahdi; Dale G. Nagle; Yu-Dong Zhou

The transcription factor hypoxia-inducible factor-1 (HIF-1) represents an important molecular target for anticancer drug discovery. In a T47D cell-based reporter assay, the Caulerpa spp. algal pigment caulerpin (1) inhibited hypoxia-induced as well as 1,10-phenanthroline-induced HIF-1 activation. The angiogenic factor vascular endothelial growth factor (VEGF) is regulated by HIF-1. Caulerpin (10 microM) suppressed hypoxic induction of secreted VEGF protein and the ability of hypoxic T47D cell-conditioned media to promote tumor angiogenesis in vitro. Under hypoxic conditions, 1 (10 microM) blocked the induction of HIF-1alpha protein, the oxygen-regulated subunit that controls HIF-1 activity. Reactive oxygen species produced by mitochondrial complex III are believed to act as a signal of cellular hypoxia that leads to HIF-1alpha protein induction and activation. Further mechanistic studies revealed that 1 inhibits mitochondrial respiration at electron transport chain (ETC) complex I (NADH-ubiquinone oxidoreductase). Under hypoxic conditions, it is proposed that 1 may disrupt mitochondrial ROS-regulated HIF-1 activation and HIF-1 downstream target gene expression by inhibiting the transport or delivery of electrons to complex III.


Tetrahedron Letters | 1995

Nakienones A-C and nakitriol, new cytotoxic cyclic C11 metabolites from an okinawan cyanobacterial (Synechocystis sp.) overgrowth of coral

Dale G. Nagle; William H. Gerwick

Abstract Nakienones A-C and nakitriol, a series of reactive cytotoxic metabolites, were isolated from dead and necrotic branches of stony coral ( Acropora sp.) which were completely covered with a gray-black mat of cyanobacteria ( Synechocystis sp.). Their structures were determined spectroscopically by interpretation of 2D-NMR experiments, including HMBC and NOESY, and by comparison with model compounds.

Collaboration


Dive into the Dale G. Nagle's collaboration.

Top Co-Authors

Avatar

Yu-Dong Zhou

University of Mississippi

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fakhri Mahdi

University of Mississippi

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yang Liu

University of Mississippi

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lin Du

University of Mississippi

View shared research outputs
Top Co-Authors

Avatar

Ikhlas A. Khan

University of Mississippi

View shared research outputs
Top Co-Authors

Avatar

J. Brian Morgan

University of Mississippi

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge