101
|
Altmeyer MA, Marschner A, Schiffmann R, Klein CD. Subtype-selectivity of metal-dependent methionine aminopeptidase inhibitors. Bioorg Med Chem Lett 2010; 20:4038-44. [PMID: 20621724 DOI: 10.1016/j.bmcl.2010.05.093] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 05/21/2010] [Accepted: 05/22/2010] [Indexed: 11/26/2022]
Abstract
Inhibitors of methionine aminopeptidases (MetAPs) are treatment options for various pathological conditions. Several inhibitor classes have been described previously, but only few data on the subtype selectivity, which is of crucial importance for these enzymes, is available. We present a systematic study on the subtype- and species-selectivity of MetAP inhibitors that require the binding of an auxiliary metal ion. This includes, in particular, compounds based on the benzimidazole pharmacophore, but also hydroxyquinoline and picolinic acid derivatives. Our data indicates that a significant degree of selectivity can be attained with metal-dependent MetAP inhibitors.
Collapse
Affiliation(s)
- Markus A Altmeyer
- Department of Medicinal Chemistry, Universität Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
102
|
Mucha A, Drag M, Dalton JP, Kafarski P. Metallo-aminopeptidase inhibitors. Biochimie 2010; 92:1509-29. [PMID: 20457213 PMCID: PMC7117057 DOI: 10.1016/j.biochi.2010.04.026] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 04/29/2010] [Indexed: 01/05/2023]
Abstract
Aminopeptidases are enzymes that selectively hydrolyze an amino acid residue from the N-terminus of proteins and peptides. They are important for the proper functioning of prokaryotic and eukaryotic cells, but very often are central players in the devastating human diseases like cancer, malaria and diabetes. The largest aminopeptidase group include enzymes containing metal ion(s) in their active centers, which often determines the type of inhibitors that are the most suitable for them. Effective ligands mostly bind in a non-covalent mode by forming complexes with the metal ion(s). Here, we present several approaches for the design of inhibitors for metallo-aminopeptidases. The optimized structures should be considered as potential leads in the drug discovery process against endogenous and infectious diseases.
Collapse
Affiliation(s)
- Artur Mucha
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland.
| | | | | | | |
Collapse
|
103
|
Abstract
Protein degradation is the cell's mechanism of eliminating misfolded or unwanted proteins. The pathway by which proteins are degraded occurs through the ubiquitin-proteasome system. Ubiquitin is a small 9-kD (kDa) protein that is attached to proteins. A minimum of four ubiquitins are required for proteins to be recognized by the degradation machinery, known as the 26S proteasome. Defects in ubiquitination have been identified in a number of diseases, including cancer, neurodegenerative diseases, and metabolic disorders. We sought to exploit the delicate balance between protein synthesis and degradation to treat cancer by designing a chimeric molecule, known as Protac (Proteolysis Targeting Chimeric molecule). Protacs are heterobifunctional nanomolecules that are approximately 10 nm in size and can recruit proteins that cause cancer to the ubiquitin-proteasome machinery for degradation. In this review, we discuss the development of this novel technology for the treatment of cancer.
Collapse
Affiliation(s)
- Kathleen M Sakamoto
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA.
| |
Collapse
|
104
|
Yasuda C, Sakata S, Kakinoki S, Takeyama Y, Ohyanagi H, Shiozaki H. In vivo evaluation of microspheres containing the angiogenesis inhibitor, TNP-470, and the metastasis suppression with liver metastatic model implanted neuroblastoma. PATHOPHYSIOLOGY 2010; 17:149-55. [DOI: 10.1016/j.pathophys.2009.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 04/14/2009] [Accepted: 04/14/2009] [Indexed: 10/20/2022] Open
|
105
|
Livingstone M, Larsson O, Sukarieh R, Pelletier J, Sonenberg N. A chemical genetic screen for mTOR pathway inhibitors based on 4E-BP-dependent nuclear accumulation of eIF4E. ACTA ACUST UNITED AC 2010; 16:1240-9. [PMID: 20064434 DOI: 10.1016/j.chembiol.2009.11.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 10/20/2009] [Accepted: 11/13/2009] [Indexed: 12/31/2022]
Abstract
The signal transduction pathway wherein mTOR regulates cellular growth and proliferation is an active target for drug discovery. The search for new mTOR inhibitors has recently yielded a handful of promising compounds that hold therapeutic potential. This search has been limited by the lack of a high-throughput assay to monitor the phosphorylation of a direct rapamycin-sensitive mTOR substrate in cells. Here we describe a novel cell-based chemical genetic screen useful for efficiently monitoring mTOR signaling to 4E-BPs in response to stimuli. The screen is based on the nuclear accumulation of eIF4E, which occurs in a 4E-BP-dependent manner specifically upon inhibition of mTOR signaling. Using this assay in a small-scale screen, we have identified several compounds not previously known to inhibit mTOR signaling, demonstrating that this method can be adapted to larger screens.
Collapse
Affiliation(s)
- Mark Livingstone
- Department of Biochemistry and McGill Cancer Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | | | | | | | | |
Collapse
|
106
|
|
107
|
Olaleye O, Raghunand TR, Bhat S, He J, Tyagi S, Lamichhane G, Gu P, Zhou J, Zhang Y, Grosset J, Bishai WR, Liu JO. Methionine aminopeptidases from Mycobacterium tuberculosis as novel antimycobacterial targets. CHEMISTRY & BIOLOGY 2010; 17:86-97. [PMID: 20142044 PMCID: PMC3165048 DOI: 10.1016/j.chembiol.2009.12.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 12/09/2009] [Accepted: 12/28/2009] [Indexed: 10/19/2022]
Abstract
Methionine aminopeptidase (MetAP) is a metalloprotease that removes the N-terminal methionine during protein synthesis. To assess the importance of the two MetAPs in Mycobacterium tuberculosis, we overexpressed and purified each of the MetAPs to near homogeneity and showed that both were active as MetAP enzymes in vitro. We screened a library of 175,000 compounds against MtMetAP1c and identified 2,3-dichloro-1,4-naphthoquinone class of compounds as inhibitors of both MtMetAPs. It was found that the MtMetAP inhibitors were active against replicating and aged nongrowing M. tuberculosis. Overexpression of either MtMetAP1a or MtMetAP1c in M. tuberculosis conferred resistance of bacterial cells to the inhibitors. Moreover, knockdown of MtMetAP1a, but not MtMetAP1c, resulted in decreased viability of M. tuberculosis. These results suggest that MtMetAP1a is a promising target for developing antituberculosis agents.
Collapse
Affiliation(s)
- Omonike Olaleye
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX 77004, USA
| | - Tirumalai R. Raghunand
- Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Shridhar Bhat
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Jian He
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Sandeep Tyagi
- Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Gyanu Lamichhane
- Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Peihua Gu
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jiangbing Zhou
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jacques Grosset
- Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - William R. Bishai
- Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Jun O. Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
108
|
Arico-Muendel CC, Benjamin DR, Caiazzo TM, Centrella PA, Contonio BD, Cook CM, Doyle EG, Hannig G, Labenski MT, Searle LL, Lind K, Morgan BA, Olson G, Paradise CL, Self C, Skinner SR, Sluboski B, Svendsen JL, Thompson CD, Westlin W, White KF. Carbamate analogues of fumagillin as potent, targeted inhibitors of methionine aminopeptidase-2. J Med Chem 2010; 52:8047-56. [PMID: 19929003 DOI: 10.1021/jm901260k] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Inhibition of methionine aminopeptidase-2 (MetAP2) represents a novel approach to antiangiogenic therapy. We describe the synthesis and activity of fumagillin analogues that address the pharmacokinetic and safety liabilities of earlier candidates in this compound class. Two-step elaboration of fumagillol with amines yielded a diverse series of carbamates at C6 of the cyclohexane spiroepoxide. The most potent of these compounds exhibited subnanomolar inhibition of cell proliferation in HUVEC and BAEC assays. Although a range of functionalities were tolerated at this position, alpha-trisubstituted amines possessed markedly decreased inhibitory activity, and this could be rationalized by modeling based on the known fumagillin-MetAP2 crystal structure. The lead compound resulting from these studies, (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-enyl)oxiran-2-yl)-1-oxaspiro[2.5]octan-6-yl (R)-1-amino-3-methyl-1-oxobutan-2-ylcarbamate, (PPI-2458), demonstrated an improved pharmacokinetic profile relative to the earlier clinical candidate TNP-470, and has advanced into phase I clinical studies in non-Hodgkin's lymphoma and solid cancers.
Collapse
|
109
|
|
110
|
Cheng KW, Wong CC, Wang M, He QY, Chen F. Identification and characterization of molecular targets of natural products by mass spectrometry. MASS SPECTROMETRY REVIEWS 2010; 29:126-155. [PMID: 19319922 DOI: 10.1002/mas.20235] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Natural products, and their derivatives and mimics, have contributed to the development of important therapeutics to combat diseases such as infections and cancers over the past decades. The value of natural products to modern drug discovery is still considerable. However, its development is hampered by a lack of a mechanistic understanding of their molecular action, as opposed to the emerging molecule-targeted therapeutics that are tailored to a specific protein target(s). Recent advances in the mass spectrometry-based proteomic approaches have the potential to offer unprecedented insights into the molecular action of natural products. Chemical proteomics is established as an invaluable tool for the identification of protein targets of natural products. Small-molecule affinity selection combined with mass spectrometry is a successful strategy to "fish" cellular targets from the entire proteome. Mass spectrometry-based profiling of protein expression is also routinely employed to elucidate molecular pathways involved in the therapeutic and possible toxicological responses upon treatment with natural products. In addition, mass spectrometry is increasingly utilized to probe structural aspects of natural products-protein interactions. Limited proteolysis, photoaffinity labeling, and hydrogen/deuterium exchange in conjunction with mass spectrometry are sensitive and high-throughput strategies that provide low-resolution structural information of non-covalent natural product-protein complexes. In this review, we provide an overview on the applications of mass spectrometry-based techniques in the identification and characterization of natural product-protein interactions, and we describe how these applications might revolutionize natural product-based drug discovery.
Collapse
Affiliation(s)
- Ka-Wing Cheng
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | | | | | | | | |
Collapse
|
111
|
ALVARADO JOHNJEFF, NEMKAL ANJANA, SAUDER JMICHAEL, RUSSELL MARIJANE, AKIYOSHI DONNAE, SHI WUXIAN, ALMO STEVENC, WEISS LOUISM. Structure of a microsporidian methionine aminopeptidase type 2 complexed with fumagillin and TNP-470. Mol Biochem Parasitol 2009; 168:158-67. [PMID: 19660503 PMCID: PMC2759695 DOI: 10.1016/j.molbiopara.2009.07.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 07/21/2009] [Accepted: 07/26/2009] [Indexed: 01/14/2023]
Abstract
Microsporidia are protists that have been reported to cause infections in both vertebrates and invertebrates. They have emerged as human pathogens particularly in patients that are immunosuppressed and cases of gastrointestinal infection, encephalitis, keratitis, sinusitis, myositis and disseminated infection are well described in the literature. While benzimidazoles are active against many species of microsporidia, these drugs do not have significant activity against Enterocytozoon bieneusi. Fumagillin and its analogues have been demonstrated to have activity invitro and in animal models of microsporidiosis and human infections due to E. bieneusi. Fumagillin and its analogues inhibit methionine aminopeptidase type 2. Encephalitozoon cuniculi MetAP2 (EcMetAP2) was cloned and expressed as an active enzyme using a baculovirus system. The crystal structure of EcMetAP2 was determined with and without the bound inhibitors fumagillin and TNP-470. This structure classifies EcMetAP2 as a member of the MetAP2c family. The EcMetAP2 structure was used to generate a homology model of the E. bieneusi MetAP2. Comparison of microsporidian MetAP2 structures with human MetAP2 provides insights into the design of inhibitors that might exhibit specificity for microsporidian MetAP2.
Collapse
Affiliation(s)
- JOHN JEFF ALVARADO
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - ANJANA NEMKAL
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | - DONNA E. AKIYOSHI
- Division of Infectious Diseases, Tufts University School of Veterinary Medicine, Grafton, MA, USA
| | - WUXIAN SHI
- Center for Synchrotron Biosciences, Case Western Reserve University, Cleveland, OH, USA
| | - STEVEN C. ALMO
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - LOUIS M. WEISS
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
112
|
Olaleye OA, Bishai WR, Liu JO. Targeting the role of N-terminal methionine processing enzymes in Mycobacterium tuberculosis. Tuberculosis (Edinb) 2009; 89 Suppl 1:S55-9. [DOI: 10.1016/s1472-9792(09)70013-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
113
|
Frottin F, Espagne C, Traverso JA, Mauve C, Valot B, Lelarge-Trouverie C, Zivy M, Noctor G, Meinnel T, Giglione C. Cotranslational proteolysis dominates glutathione homeostasis to support proper growth and development. THE PLANT CELL 2009; 21:3296-314. [PMID: 19855051 PMCID: PMC2782297 DOI: 10.1105/tpc.109.069757] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 09/17/2009] [Accepted: 10/05/2009] [Indexed: 05/18/2023]
Abstract
The earliest proteolytic event affecting most proteins is the excision of the initiating Met (NME). This is an essential and ubiquitous cotranslational process tightly regulated in all eukaryotes. Currently, the effects of NME on unknown complex cellular networks and the ways in which its inhibition leads to developmental defects and cell growth arrest remain poorly understood. Here, we provide insight into the earliest molecular mechanisms associated with the inhibition of the NME process in Arabidopsis thaliana. We demonstrate that the developmental defects induced by NME inhibition are caused by an increase in cellular proteolytic activity, primarily induced by an increase in the number of proteins targeted for rapid degradation. This deregulation drives, through the increase of the free amino acids pool, a perturbation of the glutathione homeostasis, which corresponds to the earliest limiting, reversible step promoting the phenotype. We demonstrate that these effects are universally conserved and that the reestablishment of the appropriate glutathione status restores growth and proper development in various organisms. Finally, we describe a novel integrated model in which NME, protein N-alpha-acylation, proteolysis, and glutathione homeostasis operate in a sequentially regulated mechanism that directs both growth and development.
Collapse
Affiliation(s)
- Frédéric Frottin
- Centre National de la Recherche Scientifique, Institut des Sciences du Végétal, Unité Propre de Recherche2355, Protein Maturation, Cell Fate, and Therapeutics, F-91198 Gif-sur-Yvette cedex, France
| | - Christelle Espagne
- Centre National de la Recherche Scientifique, Institut des Sciences du Végétal, Unité Propre de Recherche2355, Protein Maturation, Cell Fate, and Therapeutics, F-91198 Gif-sur-Yvette cedex, France
| | - José A. Traverso
- Centre National de la Recherche Scientifique, Institut des Sciences du Végétal, Unité Propre de Recherche2355, Protein Maturation, Cell Fate, and Therapeutics, F-91198 Gif-sur-Yvette cedex, France
| | - Caroline Mauve
- Université Paris-Sud, Institut Fédératif de Recherche87, Institut de Biotechnologie des Plantes, Plateforme Métabolisme-Métabolome, F-91405 Orsay, France
- Centre National de la Recherche Scientifique, Institut Fédératif de Recherche87, Institut de Biotechnologie des Plantes, Plateforme Métabolisme-Métabolome, Unité Mixte de Recherche 8618, F-91405 Orsay, France
| | - Benoît Valot
- Université Paris-Sud, Plateforme de Protéomique, Institut Fédératif de Recherche87, Centre National de la Recherche Scientifique/Université Paris-Sud/Institut National de la Recherche Agronomique, F-91190 Gif-sur-Yvette, France
- Centre National de la Recherche Scientifique, Plateforme de Protéomique, Institut Fédératif de Recherche87, F-91190 Gif-sur-Yvette, France
- Institut National de la Recherche Agronomique, Plateforme de Protéomique, Institut Fédératif de Recherche87, F-91190 Gif-sur-Yvette, France
| | - Caroline Lelarge-Trouverie
- Université Paris-Sud, Institut Fédératif de Recherche87, Institut de Biotechnologie des Plantes, Plateforme Métabolisme-Métabolome, F-91405 Orsay, France
- Centre National de la Recherche Scientifique, Institut Fédératif de Recherche87, Institut de Biotechnologie des Plantes, Plateforme Métabolisme-Métabolome, Unité Mixte de Recherche 8618, F-91405 Orsay, France
| | - Michel Zivy
- Université Paris-Sud, Plateforme de Protéomique, Institut Fédératif de Recherche87, Centre National de la Recherche Scientifique/Université Paris-Sud/Institut National de la Recherche Agronomique, F-91190 Gif-sur-Yvette, France
- Centre National de la Recherche Scientifique, Plateforme de Protéomique, Institut Fédératif de Recherche87, F-91190 Gif-sur-Yvette, France
- Institut National de la Recherche Agronomique, Plateforme de Protéomique, Institut Fédératif de Recherche87, F-91190 Gif-sur-Yvette, France
| | - Graham Noctor
- Université Paris-Sud, Institut Fédératif de Recherche87, Institut de Biotechnologie des Plantes, Plateforme Métabolisme-Métabolome, F-91405 Orsay, France
- Centre National de la Recherche Scientifique, Institut Fédératif de Recherche87, Institut de Biotechnologie des Plantes, Plateforme Métabolisme-Métabolome, Unité Mixte de Recherche 8618, F-91405 Orsay, France
| | - Thierry Meinnel
- Centre National de la Recherche Scientifique, Institut des Sciences du Végétal, Unité Propre de Recherche2355, Protein Maturation, Cell Fate, and Therapeutics, F-91198 Gif-sur-Yvette cedex, France
| | - Carmela Giglione
- Centre National de la Recherche Scientifique, Institut des Sciences du Végétal, Unité Propre de Recherche2355, Protein Maturation, Cell Fate, and Therapeutics, F-91198 Gif-sur-Yvette cedex, France
- Address correspondence to
| |
Collapse
|
114
|
Abstract
The medical and pharmaceutical communities are facing a dire need for new druggable targets, while, paradoxically, the targets of some drugs that are in clinical use or development remain elusive. Many compounds have been found to be more promiscuous than originally anticipated, which can potentially lead to side effects, but which may also open up additional medical uses. As we move toward systems biology and personalized medicine, comprehensively determining small molecule-target interaction profiles and mapping these on signaling and metabolic pathways will become increasingly necessary. Chemical proteomics is a powerful mass spectrometry-based affinity chromatography approach for identifying proteome-wide small molecule-protein interactions. Here we will provide a critical overview of the basic concepts and recent advances in chemical proteomics and review recent applications, with a particular emphasis on kinase inhibitors and natural products.
Collapse
Affiliation(s)
- Uwe Rix
- Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| | | |
Collapse
|
115
|
Doddareddy MR, van Westen GJP, van der Horst E, Peironcely JE, Corthals F, Ijzerman AP, Emmerich M, Jenkins JL, Bender A. Chemogenomics: Looking at biology through the lens of chemistry. Stat Anal Data Min 2009. [DOI: 10.1002/sam.10046] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
116
|
Arico-Muendel C, Centrella PA, Contonio BD, Morgan BA, O'Donovan G, Paradise CL, Skinner SR, Sluboski B, Svendsen JL, White KF, Debnath A, Gut J, Wilson N, McKerrow JH, DeRisi JL, Rosenthal PJ, Chiang PK. Antiparasitic activities of novel, orally available fumagillin analogs. Bioorg Med Chem Lett 2009; 19:5128-31. [PMID: 19648008 PMCID: PMC2745105 DOI: 10.1016/j.bmcl.2009.07.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 06/20/2009] [Accepted: 07/02/2009] [Indexed: 11/26/2022]
Abstract
Fumagillin, an irreversible inhibitor of MetAP2, has been shown to potently inhibit growth of malaria parasites in vitro. Here, we demonstrate activity of fumagillin analogs with an improved pharmacokinetic profile against malaria parasites, trypanosomes, and amoebas. A subset of the compounds showed efficacy in a murine malaria model. The observed SAR forms a basis for further optimization of fumagillin based inhibitors against parasitic targets by inhibition of MetAP2.
Collapse
|
117
|
Datta B. Roles of P67/MetAP2 as a tumor suppressor. Biochim Biophys Acta Rev Cancer 2009; 1796:281-92. [PMID: 19716858 DOI: 10.1016/j.bbcan.2009.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 08/12/2009] [Accepted: 08/22/2009] [Indexed: 12/17/2022]
Abstract
A precise balance between growth promoting signals and growth inhibitory signals plays important roles in the maintenance of healthy mammalian cells. Any deregulation of this critical balance converts normal cells into abnormal or cancerous cells. Several macromolecules are being identified and characterized that are involved in the regulation of cell signaling pathways that connect to the cell cycle and thus they play roles as tumor promoters or tumor suppressors. In situ tumor formation needs active angiogenesis, a process that generates new blood vessels from existing ones either by splitting or sprouting. Several small molecule inhibitors and proteins have been identified as inhibitors of angiogenesis. One such protein, p67/MetAP2 also known as methionine aminopeptidase 2 (MetAP2), has been shown to bind covalently to fumagillin and its derivatives that have anti-angiogenic activity. In addition to fumagillin or its derivatives, several other small molecule inhibitors of p67/MetAP2 have been recently identified and some of these drugs are in phase III trials for cancer therapy. Although molecular details of actions toward tumor suppression by these drugs are largely unknown, a significant progress has been made to understand the structure-function relationship of p67/MetAP2 and its roles in the maintenance of the levels of phosphorylation of the proportional, variant-subunit of eukaryotic initiation factor 2 (eIF2 proportional, variant) and extracellular signal-regulated kinases 1 and 2 (ERK1/2). In this article, roles of p67/MetAP2 in the suppression of cancer development are also discussed.
Collapse
Affiliation(s)
- Bansidhar Datta
- Department of Chemistry, Kent State University, Kent, OH 44242, USA.
| |
Collapse
|
118
|
Sawanyawisuth K, Wongkham C, Pairojkul C, Saeseow OT, Riggins GJ, Araki N, Wongkham S. Methionine aminopeptidase 2 over-expressed in cholangiocarcinoma: potential for drug target. Acta Oncol 2009; 46:378-85. [PMID: 17450475 DOI: 10.1080/02841860600871061] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Methionine aminopeptidases (MetAP) are proteases which remove the N-terminal methionine from newly synthesized proteins. Associations of MetAP2 with tumor progression of different cancers have been repeatedly reported. We aim to determine if MetAP2 is expressed in cholangiocarcinomas (CCA) and investigate to see if it would be a useful therapeutic target. We evaluated MetAP2 expression by immunohistochemistry in 82 patients of intrahepatic CCA. MetAP2 was expressed in bile ducts to various degrees. It was occasionally expressed with weak staining in normal bile duct epithelium but was strikingly over-expressed in dysplastic bile duct epithelia, primary and metastatic CCA tissues (p < 0.001). The increased expression of MetAP2 in proliferating bile duct was evident. All metastatic tumors had stronger expression of MetAP2 than the corresponding primary tumors. Fumagillin, a MetAP2 specific inhibitor, significantly inhibited cell proliferation in dose dependent manner and the degree of growth inhibition was dependent on the amount of cellular enzyme. The present study highlights the involvement of MetAP2 in an early event of carcinogenesis of CCA. The findings represent the first description of increased MetAP2 expression in CCA. The inhibition of enzyme activity using MetAP2 inhibitors may be a potential strategy for long-term control of tumor development and progression in CCA patients.
Collapse
Affiliation(s)
- Kanlayanee Sawanyawisuth
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | | | | | | | | | | | | |
Collapse
|
119
|
Hou L, Mori D, Takase Y, Meihua P, Kai K, Tokunaga O. Fumagillin inhibits colorectal cancer growth and metastasis in mice: in vivo and in vitro study of anti-angiogenesis. Pathol Int 2009; 59:448-461. [PMID: 19563408 DOI: 10.1111/j.1440-1827.2009.02393.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Fumagillin is an inhibitor of type 2 methionine aminopeptidase that can block blood vessel formation, but its molecular mechanism and therapeutic value in colon cancer still remain to be elucidated. In this study, male severe combined immunodeficiency (SCID) mice were injected with colon cancer cells in the subcutis and then treated with Fumagillin and Cyclo (Arg-Gly-Asp-D-Phe-Val), an integrin alphavbeta(3) antagonist. The tumor weight, microvessel density (MVD), and number of pulmonary metastatic foci were examined. Gene expression profiles were examined by microarray analysis of human umbilical endothelial cells (HUVEC). The Fumagillin-treated mice had smaller tumor mass, fewer pulmonary metastases, and lower MVD-CD105 levels than control animals. In vitro proliferation and tube formation of HUVEC was also significantly decreased by Fumagillin. Microarray analysis of Fumagillin-treated HUVEC showed upregulation of 71 genes and downregulation of 143 genes. Expression changes were involved in cell proliferation, migration, adhesion, and gene transcription. Quantitative real-time-polymerase chain reaction and western blotting showed decreased expression of cyclin E2, activated leukocyte cell adhesion molecule (ALCAM), and intercellular adhesion molecule-1 (ICAM-1) genes in the presence of Fumagillin. This downregulation by Fumagillin may be involved in the anti-angiogenesis by Fumagillin. In conclusion, Fumagillin was found to suppress colorectal cancer growth and metastasis by suppressing angiogenesis.
Collapse
Affiliation(s)
- Li Hou
- Department of Pathology and Biodefense, Faculty of Medicine, Saga University, Saga, Japan
| | | | | | | | | | | |
Collapse
|
120
|
Hartley M, Yong W, Bennett B. Heterologous expression and purification of Vibrio proteolyticus (Aeromonas proteolytica) aminopeptidase: a rapid protocol. Protein Expr Purif 2009; 66:91-101. [PMID: 19233285 PMCID: PMC2669716 DOI: 10.1016/j.pep.2009.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 02/11/2009] [Accepted: 02/11/2009] [Indexed: 11/27/2022]
Abstract
Metalloaminopeptidases (mAPs) are enzymes that are involved in HIV infectivity, tumor growth and metastasis, angiogenesis, and bacterial infection. Investigation of structure-function relationships in mAPs is a prerequisite to rational design of anti-mAP chemotherapeutics. The most intensively studied member of the biomedically important dinuclear mAPs is the prototypical secreted Vibrio proteolyticus di-zinc aminopeptidase (VpAP). The wild-type enzyme is readily purified from the supernatant of cultures of V. proteolyticus, but recombinant variants require expression in Escherichia coli. A greatly improved system for the purification of recombinant VpAP is described. A VpAP-(His)(6) polypeptide, containing an N-terminal propeptide, and a C-terminal (His)(6) adduct, was purified by metal ion affinity chromatography from the supernatant of cultures of E. coli. This single step replaced the sequence of (NH(4))(2)SO(4) fractionation, and anion-exchange and hydrophobic interaction chromatographic separations of earlier methods. Traditionally, recombinant VpAP proenzyme has been treated with proteinase K and with heat (70 degrees C), to remove the N- and C-terminal regions, and yield the mature active enzyme. This method is unsuitable for VpAP variants that are unstable towards these treatments. In the new method, the hitherto noted, but not fully appreciated, ability of VpAP to autocatalyze the hydrolysis of the N-terminal propeptide and C-terminal regions was exploited; extensive dialysis of the highly purified VpAP-(His)(6) full-length polypeptide yielded the mature active protein without recourse to proteinase K or heat treatment. Purification of variants that have previously defied isolation as mature forms of the protein was thus carried out.
Collapse
Affiliation(s)
- Mariam Hartley
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226-0509, USA
| | | | | |
Collapse
|
121
|
Chen X, Xie S, Bhat S, Kumar N, Shapiro TA, Liu JO. Fumagillin and fumarranol interact with P. falciparum methionine aminopeptidase 2 and inhibit malaria parasite growth in vitro and in vivo. ACTA ACUST UNITED AC 2009; 16:193-202. [PMID: 19246010 DOI: 10.1016/j.chembiol.2009.01.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 12/21/2008] [Accepted: 01/05/2009] [Indexed: 11/24/2022]
Abstract
The fumagillin family of natural products is known to inhibit angiogenesis through irreversible inhibition of human type 2 methionine aminopeptidase (MetAP2). Recently, fumagillin and TNP-470 were reported to possess antimalarial activity in vitro, and it was hypothesized that this inhibition was mediated by interaction with the putative malarial ortholog of human MetAP2. In this report, we have overexpressed and purified to near-homogeneity PfMetAP2 from bacteria, yeast, and insect cells. Although none of the recombinant forms of PfMetAP2 exhibited enzymatic activity in existing assays, PfMetAP2 proteins expressed in both yeast and insect cells were able to bind to fumagillin in a pull-down assay. The interaction between fumagillin and analogs with PfMetAP2 was further demonstrated using a newly established mammalian three-hybrid assay incorporating a conjugate between dexamethasone and fumagillin. Unlike human (Hs)MetAP2, it was found that PfMetAP2 is bound to fumagillin noncovalently. Importantly, a new analog of fumagillin, fumarranol, was demonstrated to interact with PfMetAP2 and inhibit the growth of both chloroquine-sensitive and drug-resistant Plasmodium falciparum strains in vitro. Antiparasite activity of fumagillin and fumarranol was also demonstrated in vivo using a mouse malaria model. These findings suggest that PfMetAP2 is a viable target, and fumarranol is a promising lead compound for the development of novel antimalarial agents.
Collapse
Affiliation(s)
- Xiaochun Chen
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
122
|
Mitra S, Sheppard G, Wang J, Bennett B, Holz RC. Analyzing the binding of Co(II)-specific inhibitors to the methionyl aminopeptidases from Escherichia coli and Pyrococcus furiosus. J Biol Inorg Chem 2009; 14:573-85. [PMID: 19198897 PMCID: PMC2678238 DOI: 10.1007/s00775-009-0471-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Accepted: 12/31/2008] [Indexed: 10/21/2022]
Abstract
Methionine aminopeptidases (MetAPs) represent a unique class of protease that is capable of the hydrolytic removal of an N-terminal methionine residue from nascent polypeptide chains. MetAPs are physiologically important enzymes; hence, there is considerable interest in developing inhibitors that can be used as antiangiogenic and antimicrobial agents. A detailed kinetic and spectroscopic study has been performed to probe the binding of a triazole-based inhibitor and a bestatin-based inhibitor to both Mn(II)- and Co(II)-loaded type-I (Escherichia coli) and type-II (Pyrococcus furiosus) MetAPs. Both inhibitors were found to be moderate competitive inhibitors. The triazole-type inhibitor was found to interact with both active-site metal ions, while the bestatin-type inhibitor was capable of switching its mode of binding depending on the metal in the active site and the type of MetAP enzyme.
Collapse
Affiliation(s)
- Sanghamitra Mitra
- Department of Chemistry, Loyola University-Chicago, Chicago, IL 60626, USA
| | | | | | | | | |
Collapse
|
123
|
Brahn E, Schoettler N, Lee S, Banquerigo ML. Involution of collagen-induced arthritis with an angiogenesis inhibitor, PPI-2458. J Pharmacol Exp Ther 2009; 329:615-24. [PMID: 19218530 PMCID: PMC2672877 DOI: 10.1124/jpet.108.148478] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Accepted: 02/12/2009] [Indexed: 12/12/2022] Open
Abstract
Pannus formation, in both rheumatoid arthritis (RA) and collagen-induced arthritis (CIA), is angiogenesis-dependent. PPI-2458 [(1R)-1-carbamoyl-2-methyl]-carbamic acid-(3R,3S,5S, 6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methyl-but-2-enyl)oxiranyl]-1-oxaspiro(2*5)oct-6-yl ester], a new fumagillin derivative known to inhibit methionine aminopeptidase 2 (MetAP-2) and endothelial proliferation at the late G(1) phase, was evaluated in CIA rats to study its potential to involute synovitis. Arthritic syngeneic LOU rats received either a vehicle control or various dosages of oral, intravenous, or subcutaneous PPI-2458. Plasma samples were analyzed to determine a pharmacokinetic profile of PPI-2458, and whole blood was evaluated by flow cytometry to assess the effect on lymphocyte subsets. At 15 mg/kg i.v., 30 mg/kg s.c., or 100 mg/kg p.o., there was a significant reduction in clinical severity scores (p < 0.001) and blinded radiographic scores (p < 0.001) compared with vehicle control groups. Structural damage was virtually eliminated with PPI-2458. Continuous inhibition of MetAP-2 was needed to maintain benefits, although pannus involution could be achieved with the inhibitor when escape flares occurred. Pharmacokinetic analysis after a single p.o. dose showed a rapid T(max) value of 15 min followed by biphasic elimination (t(1/2), approximately 20 min and t(1/2), approximately 5 h) and an estimated oral bioavailability of approximately 15%. Flow cytometry revealed a dose-dependent decrease in white blood cells and lymphocytes manifested as decreases in circulating CD3+ T cells and natural killer cells. PPI-2458, however, did not seem to be immunosuppressive, as determined by delayed-type hypersensitivity or IgG antibody assays. These studies indicate that the MetAP-2 inhibitor PPI-2458 can regress established CIA and that angiogenic mechanisms might be important targets in the treatment of other pannus-mediated diseases such as RA.
Collapse
MESH Headings
- Administration, Oral
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/chemistry
- Angiogenesis Inhibitors/pharmacokinetics
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Biological Availability
- Collagen Type II
- Epoxy Compounds/administration & dosage
- Epoxy Compounds/chemistry
- Epoxy Compounds/pharmacokinetics
- Epoxy Compounds/therapeutic use
- Flow Cytometry
- Injections, Intravenous
- Injections, Subcutaneous
- Molecular Structure
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Rats
- Rats, Inbred Strains
- Valine/administration & dosage
- Valine/analogs & derivatives
- Valine/chemistry
- Valine/pharmacokinetics
- Valine/therapeutic use
Collapse
Affiliation(s)
- Ernest Brahn
- Division of Rheumatology, UCLA School of Medicine, Los Angeles, CA 90095-1670, USA.
| | | | | | | |
Collapse
|
124
|
Sashidhara KV, White KN, Crews P. A selective account of effective paradigms and significant outcomes in the discovery of inspirational marine natural products. JOURNAL OF NATURAL PRODUCTS 2009; 72:588-603. [PMID: 19209899 PMCID: PMC2837139 DOI: 10.1021/np800817y] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Marine natural products continue to be a source of significant molecular structures that serve as a stimulus to seed further significant research. This account reviews some of the major advances in the study of marine biomolecules made at UC Santa Cruz over more than three decades. The continuing challenge of discovery and characterization of what we term "inspirational molecular structures" will be presented in a comprehensive fashion. Examples of privileged molecular structures and their impact on biomedicinal research will be an important theme. The three major groups of organisms explored include seaweeds, sponges, and marine-derived fungi, and the study of their active principles has greatly benefited from synergistic collaborations with both academic and biopharmaceutical groups. The concluding sections of this chronicle will touch on prospects for future outcomes involving new sources and strategies.
Collapse
Affiliation(s)
- Koneni V. Sashidhara
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064
| | - Kimberly N. White
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064
| | - Phillip Crews
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064
| |
Collapse
|
125
|
Mitra S, Bennett B, Holz RC. Mutation of H63 and its catalytic affect on the methionine aminopeptidase from Escherichia coli. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1794:137-43. [PMID: 18952013 PMCID: PMC2674292 DOI: 10.1016/j.bbapap.2008.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 09/11/2008] [Accepted: 09/13/2008] [Indexed: 05/27/2023]
Abstract
In order to gain insight into the mechanistic role of a flexible exterior loop near the active site, made up of Y62, H63, G64, and Y65, that has been proposed to play an important role in substrate binding and recognition in the methionyl aminopeptidase from Escherichia coli (EcMetAP-I), the H63A enzyme was prepared. Mutation of H63 to alanine does not affect the ability of the enzyme to bind divalent metal ions. The specific activity of H63A EcMetAP-I was determined using four different substrates of varying lengths, namely, l-Met-p-NA, MAS, MGMM and MSSHRWDW. For the smallest/shortest substrate (l-Met-p-NA) the specific activity decreased nearly seven fold but as the peptide length increased, the specific activity also increased and became comparable to WT EcMetAP-I. This decrease in specific activity is primarily due to a decrease in the observed k(cat) values, which decreases nearly sixty-fold for l-Met-p-NA while only a four-fold decrease is observed for the tri- and tetra-peptide substrates. Interestingly, no change in k(cat) was observed when the octa-peptide MSSHRWDW was used as a substrate. These data suggest that H63 affects the hydrolysis of small peptide substrates whereas large peptides can overcome the observed loss in binding energy, as predicted from K(m) values, by additional hydrophilic and hydrophobic interactions.
Collapse
Affiliation(s)
- Sanghamitra Mitra
- Department of Chemistry, Boston University, Boston, MA 02215-2521, USA
| | | | | |
Collapse
|
126
|
Ren YR, Pan F, Parvez S, Fleig A, Chong CR, Xu J, Dang Y, Zhang J, Jiang H, Penner R, Liu JO. Clofazimine inhibits human Kv1.3 potassium channel by perturbing calcium oscillation in T lymphocytes. PLoS One 2008; 3:e4009. [PMID: 19104661 PMCID: PMC2602975 DOI: 10.1371/journal.pone.0004009] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2008] [Accepted: 11/08/2008] [Indexed: 01/03/2023] Open
Abstract
The Kv1.3 potassium channel plays an essential role in effector memory T cells and has been implicated in several important autoimmune diseases including multiple sclerosis, psoriasis and type 1 diabetes. A number of potent small molecule inhibitors of Kv1.3 channel have been reported, some of which were found to be effective in various animal models of autoimmune diseases. We report herein the identification of clofazimine, a known anti-mycobacterial drug, as a novel inhibitor of human Kv1.3. Clofazimine was initially identified as an inhibitor of intracellular T cell receptor-mediated signaling leading to the transcriptional activation of human interleukin-2 gene in T cells from a screen of the Johns Hopkins Drug Library. A systematic mechanistic deconvolution revealed that clofazimine selectively blocked the Kv1.3 channel activity, perturbing the oscillation frequency of the calcium-release activated calcium channel, which in turn led to the inhibition of the calcineurin-NFAT signaling pathway. These effects of clofazimine provide the first line of experimental evidence in support of a causal relationship between Kv1.3 and calcium oscillation in human T cells. Furthermore, clofazimine was found to be effective in blocking human T cell-mediated skin graft rejection in an animal model in vivo. Together, these results suggest that clofazimine is a promising immunomodulatory drug candidate for treating a variety of autoimmune disorders.
Collapse
Affiliation(s)
- Yunzhao R. Ren
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Program in Biochemistry, Cellular and Molecular Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Fan Pan
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Suhel Parvez
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, Hawaii, United States of America
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, Hawaii, United States of America
| | - Curtis R. Chong
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jing Xu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yongjun Dang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jin Zhang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Hongsi Jiang
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, Illinois, United States of America
| | - Reinhold Penner
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, Hawaii, United States of America
| | - Jun O. Liu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Program in Biochemistry, Cellular and Molecular Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
127
|
Priest RC, Spaull J, Buckton J, Grimley RL, Sims M, Binks M, Malhotra R. Immunomodulatory activity of a methionine aminopeptidase-2 inhibitor on B cell differentiation. Clin Exp Immunol 2008; 155:514-22. [PMID: 19068103 DOI: 10.1111/j.1365-2249.2008.03843.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Methionine aminopeptidase-2 (MetAP-2) inhibitors have potent anti-angiogenesis activity and are being developed for the treatment of solid tumours. The recently observed specific expression of MetAP-2 in germinal centre B cells suggests that it has a role in regulating B cell function. We have demonstrated a potent MetAP-2-dependent inhibitory effect on the antibody secretion from B cell receptor and CD40 co-stimulated primary human B cells in the presence of interleukin-21. The effect of MetAP-2 inhibition on antibody secretion was due to a block in differentiation of B cells into plasma cells. Immunohistochemical analysis of germinal centres from human, mouse and marmoset spleen showed a similar expression pattern of MetAP-2 in the marmoset and man, whereas mouse spleen showed no detectable expression. In a marmoset, T dependent immunization model, the MetAP-2 inhibitor suppressed an antigen-specific antibody response. Furthermore, histological analysis showed loss of B cells in the spleen and disrupted germinal centre formation. These results provide experimental evidence to support a novel role for MetAP-2 in immunomodulation. These effects of MetAP-2 are mediated by disruption of the germinal centre reaction and a block in the differentiation of B cells into plasma cells.
Collapse
Affiliation(s)
- R C Priest
- II CEDD, GlaxoSmithKline, Gunnel's Wood Road, Stevenage, Hertfordshire, UK.
| | | | | | | | | | | | | |
Collapse
|
128
|
Mitra S, Job KM, Meng L, Bennett B, Holz RC. Analyzing the catalytic role of Asp97 in the methionine aminopeptidase from Escherichia coli. FEBS J 2008; 275:6248-59. [PMID: 19019076 PMCID: PMC2699115 DOI: 10.1111/j.1742-4658.2008.06749.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An active site aspartate residue, Asp97, in the methionine aminopeptidase (MetAPs) from Escherichia coli (EcMetAP-I) was mutated to alanine, glutamate, and asparagine. Asp97 is the lone carboxylate residue bound to the crystallographically determined second metal-binding site in EcMetAP-I. These mutant EcMetAP-I enzymes have been kinetically and spectroscopically characterized. Inductively coupled plasma-atomic emission spectroscopy analysis revealed that 1.0 +/- 0.1 equivalents of cobalt were associated with each of the Asp97-mutated EcMetAP-Is. The effect on activity after altering Asp97 to alanine, glutamate or asparagine is, in general, due to a approximately 9000-fold decrease in k(ca) towards Met-Gly-Met-Met as compared to the wild-type enzyme. The Co(II) d-d spectra for wild-type, D97E and D97A EcMetAP-I exhibited very little difference in form, in each case, between the monocobalt(II) and dicobalt(II) EcMetAP-I, and only a doubling of intensity was observed upon addition of a second Co(II) ion. In contrast, the electronic absorption spectra of [Co_(D97N EcMetAP-I)] and [CoCo(D97N EcMetAP-I)] were distinct, as were the EPR spectra. On the basis of the observed molar absorptivities, the Co(II) ions binding to the D97E, D97A and D97N EcMetAP-I active sites are pentacoordinate. Combination of these data suggests that mutating the only nonbridging ligand in the second divalent metal-binding site in MetAPs to an alanine, which effectively removes the ability of the enzyme to form a dinuclear site, provides a MetAP enzyme that retains catalytic activity, albeit at extremely low levels. Although mononuclear MetAPs are active, the physiologically relevant form of the enzyme is probably dinuclear, given that the majority of the data reported to date are consistent with weak cooperative binding.
Collapse
Affiliation(s)
- Sanghamitra Mitra
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, USA
| | | | | | | | | |
Collapse
|
129
|
Watterson SJ, Mitra S, Swierczek SI, Bennett B, Holz RC. Kinetic and spectroscopic analysis of the catalytic role of H79 in the methionine aminopeptidase from Escherichia coli. Biochemistry 2008; 47:11885-93. [PMID: 18855426 PMCID: PMC2720795 DOI: 10.1021/bi801499g] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To gain insight into the role of the strictly conserved histidine residue, H79, in the reaction mechanism of the methionyl aminopeptidase from Escherichia coli ( EcMetAP-I), the H79A mutated enzyme was prepared. Co(II)-loaded H79A exhibits an overall >7000-fold decrease in specific activity. The almost complete loss of activity is primarily due to a >6000-fold decrease in k cat. Interestingly, the K m value obtained for Co(II)-loaded H79A was approximately half the value observed for wild-type (WT) EcMetAP-I. Consequently, k cat/ K m values decreased only 3000-fold. On the other hand, the observed specific activity of Mn(II)-loaded H79A EcMetAP-I decreased by approximately 2.6-fold while k cat decreased by approximately 3.5-fold. The observed K m value for Mn(II)-loaded H79A EcMetAP-I was approximately 1.4-fold larger than that observed for WT EcMetAP-I, resulting in a k cat/ K m value that is lower by approximately 3.4-fold. Metal binding, UV-vis, and EPR data indicate that the active site is unperturbed by mutation of H79, as suggested by X-ray crystallographic data. Kinetic isotope data indicate that H79 does not transfer a proton to the newly forming amine since a single proton is transferred in the transition state for both the WT and H79A EcMetAP-I enzymes. Therefore, H79 functions to position the substrate by hydrogen bonding to either the amine group of the peptide linkage or a backbone carbonyl group. Together, these data provide new insight into the catalytic mechanism of EcMetAP-I.
Collapse
Affiliation(s)
- Sarah J. Watterson
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300
| | - Sanghamitra Mitra
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300
| | - Sabina I. Swierczek
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300
| | - Brian Bennett
- National Biomedical EPR Center, Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226-0509
| | - Richard C. Holz
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300
- Department of Chemistry, Loyola University, 1068 West Sheridan Road, Chicago, Illinois 60626
| |
Collapse
|
130
|
Warder SE, Tucker LA, McLoughlin SM, Strelitzer TJ, Meuth JL, Zhang Q, Sheppard GS, Richardson PL, Lesniewski R, Davidsen SK, Bell RL, Rogers JC, Wang J. Discovery, identification, and characterization of candidate pharmacodynamic markers of methionine aminopeptidase-2 inhibition. J Proteome Res 2008; 7:4807-20. [PMID: 18828628 DOI: 10.1021/pr800388p] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The catalytic activity of methionine aminopeptidase-2 (MetAP2) has been pharmacologically linked to cell growth, angiogenesis, and tumor progression, making this an attractive target for cancer therapy. An assay for monitoring specific protein changes in response to MetAP2 inhibition, allowing pharmacokinetic (PK)/pharmacodynamic (PD) models to be established, could dramatically improve clinical decision-making. Candidate MetAP2-specific protein substrates were discovered from undigested cell culture-derived proteomes by MALDI-/SELDI-MS profiling and a biochemical method using (35)S-Met labeled protein lysates. Substrates were identified either as intact proteins by FT-ICR-MS or applying in-gel protease digestions followed by LC-MS/MS. The combination of these approaches led to the discovery of novel MetAP2-specific substrates including thioredoxin-1 (Trx-1), SH3 binding glutamic acid rich-like protein (SH3BGRL), and eukaryotic elongation factor-2 (eEF2). These studies also confirmed glyceraldehye 3-phosphate dehydrogenase (GAPDH) and cyclophillin A (CypA) as MetAP2 substrates. Additional data in support of these proteins as MetAP2-specific substrates were provided by in vitro MetAP1/MetAP2 enzyme assays with the corresponding N-terminal derived peptides and 1D/2D Western analyses of cellular and tissue lysates. FT-ICR-MS characterization of all intact species of the 18 kDa substrate, CypA, enabled a SELDI-MS cell-based assay to be developed for correlating N-terminal processing and inhibition of proliferation. The MetAP2-specific protein substrates discovered in this study have diverse properties that should facilitate the development of reagents for testing in preclinical and clinical environments.
Collapse
Affiliation(s)
- Scott E Warder
- Advanced Technology and Cancer Research, Global Pharmaceutical Research and Development, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6202, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Stevanovic J, Stanimirovic Z, Radakovic M, Stojic V. In vitro evaluation of the clastogenicity of fumagillin. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2008; 49:594-601. [PMID: 18613037 DOI: 10.1002/em.20409] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Fumagillin, an antibiotic compound produced by Aspergillus fumigatus, is effective against microsporidia and various Amoeba species, but is also toxic when administered systemically to mammals. Furthermore, a recent in vivo study by Stanimirovic Z et al. 2007: (Mutat Res 628:1-10) indicated genotoxic effects of fumagillin. The aim of the present study was to investigate and explain the clastogenic effects of fumagillin (in the form of fumagillin dicyclohexylamine salt) on human peripheral blood lymphocytes in vitro by sister-chromatid exchanges (SCE), chromosome aberrations (CA), and micronucleus (MN) tests. The mitotic index (MI), proliferation index (PI), and nuclear division index (NDI) were calculated to evaluate the cytotoxic potential of fumagillin. Five concentrations of fumagillin (0.34, 0.68, 1.02, 3.07, and 9.20 microg/ml) were applied to lymphocyte cultures. All the tested concentrations of fumagillin increased the frequency of SCE per cell significantly (P < 0.001 or P < 0.01) compared with the negative control. A significant (P < 0.001) increase in frequency of structural CA was observed at the three highest concentrations in comparison with the negative control. In addition, the three highest test concentrations increased MN formation and decreased MI, PI, and NDI significantly compared with the negative control. The present results indicate that fumagillin is clastogenic and cytotoxic to cultured human lymphocytes.
Collapse
Affiliation(s)
- Jevrosima Stevanovic
- Department of Biology, Faculty of Veterinary Medicine, University of Belgrade, Belgrade, Serbia.
| | | | | | | |
Collapse
|
132
|
Chai SC, Wang WL, Ye QZ. FE(II) is the native cofactor for Escherichia coli methionine aminopeptidase. J Biol Chem 2008; 283:26879-85. [PMID: 18669631 DOI: 10.1074/jbc.m804345200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Divalent metal ions play a critical role in the removal of N-terminal methionine from nascent proteins by methionine aminopeptidase (MetAP). Being an essential enzyme for bacteria, MetAP is an appealing target for the development of novel antibacterial drugs. Although purified enzyme can be activated by several divalent metal ions, the exact metal ion used by MetAP in cells is unknown. Many MetAP inhibitors are highly potent on purified enzyme, but they fail to show significant inhibition of bacterial growth. One possibility for the failure is a disparity of the metal used in activation of purified MetAP and the metal actually used by MetAP inside bacterial cells. Therefore, the challenge is to elucidate the physiologically relevant metal for MetAP and discover MetAP inhibitors that can effectively inhibit cellular MetAP. We have recently discovered MetAP inhibitors with selectivity toward different metalloforms of Escherichia coli MetAP, and with these unique inhibitors, we characterized their inhibition of MetAP enzyme activity in a cellular environment. We observed that only inhibitors that are selective for the Fe(II)-form of MetAP were potent in this assay. Further, we found that only these Fe(II)-form selective inhibitors showed significant inhibition of growth of five E. coli strains and two Bacillus strains. We confirmed their cellular target as MetAP by analysis of N-terminal processed and unprocessed recombinant glutathione S-transferase proteins. Therefore, we conclude that Fe(II) is the likely metal used by MetAP in E. coli and other bacterial cells.
Collapse
Affiliation(s)
- Sergio C Chai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
133
|
Chen GJ, Weylie B, Hu C, Zhu J, Forough R. FGFR1/PI3K/AKT signaling pathway is a novel target for antiangiogenic effects of the cancer drug fumagillin (TNP-470). J Cell Biochem 2008; 101:1492-504. [PMID: 17295210 DOI: 10.1002/jcb.21265] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Fibroblast growth factor-1 (FGF1), a prototypic member of the FGF family, is a potent angiogenic factor. Although FGF-stimulated angiogenesis has been extensively studied, the molecular mechanisms regulating FGF1-induced angiogenesis are poorly understood in vivo. Fumagillin, an antiangiogenic fungal metabolite, has the ability to inhibit FGF-stimulated angiogenesis in the chicken chorioallantoic membrane (CAM). In the current study, chicken CAMs were transfected with a signal peptide-containing version of the FGF1 gene construct (sp-FGF1). Transfected CAMs were then analyzed in the presence and absence of fumagillin treatment with respect to the mRNA expression levels and protein activity of the FGF1 receptor protein (FGFR1), phosphatidylinositol 3-kinase (PI3K), and its immediate downstream target, AKT-1 (protein kinase B). Treatment of sp-FGF1-transfected CAMs with fumagillin showed downregulation for both PI3K and AKT-1 proteins in mRNA expression and protein activity. In contrast, no major alterations in FGFR1 mRNA expression level were observed. Similar patterns of mRNA expression for the above three proteins were observed when the CAMs were treated with recombinant FGF1 protein in place of sp-FGF1 gene transfection. Investigation using biotin-labeled fumagillin showed that only the FGF1 receptor protein containing the cytoplasmic domain demonstrated binding to fumagillin. Furthermore, we demonstrated endothelial-specificity of the proposed antiangiogenic signaling cascade using an in vitro system. Based on these findings, we conclude that the binding of fumagillin to the cytoplasmic domain of the FGF1 receptor inhibited FGF1-stimulated angiogenesis both in vitro and in vivo.
Collapse
Affiliation(s)
- Gregory J Chen
- Department of Systems Biology and Translational Medicine, College of Medicine, The Texas A&M University System Health Science Center, College Station, Texas 77843, USA
| | | | | | | | | |
Collapse
|
134
|
Wong CC, Cheng KW, He QY, Chen F. Unraveling the molecular targets of natural products: Insights from genomic and proteomic analyses. Proteomics Clin Appl 2008; 2:338-54. [DOI: 10.1002/prca.200880002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2007] [Indexed: 11/11/2022]
|
135
|
Ectopic expression of methionine aminopeptidase-2 causes cell transformation and stimulates proliferation. Oncogene 2008; 27:3967-76. [PMID: 18264137 DOI: 10.1038/onc.2008.14] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Methionine aminopeptidase-2 (MetAP2) processes N-terminal methionine from nascent cellular proteins. Inhibition of MetAP2 has been shown to block angiogenesis and suppress tumor growth in preclinical tumor models. However, the biological role of MetAP2 in cancer is not well understood. We examined the effect of three distinct chemical classes of MetAP2 inhibitors on the growth of a panel of human cancer cells in vitro. All MetAP2 inhibitors caused inhibition of tumor cell growth in both anchorage-dependent and, particularly, in anchorage-independent manner. These data prompted us to examine the possible roles of MetAP2 in cancers. Ectopic expression of MetAP2 in NIH-3T3 cells caused transformation, evidenced by the formation of foci in monolayer culture and growth of large colonies in soft agar. Overexpression of MetAP2 in an immortalized bronchial epithelial cell line NL20 accelerated growth. These phenotypes induced by the overexpression of MetAP2 were reversed by the treatment with MetAP2 inhibitors, indicating that the catalytic function of MetAP2 was essential. Accordingly, overexpression of a catalytically inactive MetAP2 resulted in growth retardation of HT1080 tumor cells, suggesting a dominant-negative role of the inactive MetAP2 mutant. Finally, we analysed the expression of MetAP2 in patient cancer samples by immunohistochemistry. Moderate-to-high staining was identified in the majority of breast, colon, lung, ovarian and prostate carcinomas examined. These data suggest that MetAP2 plays an important role in tumor cell growth and may contribute to tumorigenesis.
Collapse
|
136
|
Meinnel T, Giglione C. Tools for analyzing and predicting N-terminal protein modifications. Proteomics 2008; 8:626-49. [DOI: 10.1002/pmic.200700592] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
137
|
TNP-470: The Resurrection of the First Synthetic Angiogenesis Inhibitor. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
138
|
Vedantham P, Guerra JM, Schoenen F, Huang M, Gor PJ, Georg GI, Wang JL, Neuenswander B, Lushington GH, Mitscher LA, Ye QZ, Hanson PR. Ionic immobilization, diversification, and release: application to the generation of a library of methionine aminopeptidase inhibitors. ACTA ACUST UNITED AC 2007; 10:185-94. [PMID: 18163595 DOI: 10.1021/cc700085c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Development of an ionic immobilization, diversification, and release method for the generation of methionine aminopeptidase inhibitors is reported. This method involves the immobilization of 5-bromofuran-2-carboxylic acid and 5-bromothiophene-2-carboxylic acid onto PS-BEMP, followed by Suzuki reaction on a resin-bound intermediate and subsequent release to provide products in moderate yields and excellent purities. Compound potencies were evaluated on the Co(II), Mn(II), Ni(II), and Fe(II) forms of Escherichia coli MetAP1. The furoic-acid analogs were found to be Mn(II) selective with IC 50 values in the low micromolar range. Qualitative SAR analysis, supplemented by molecular modeling studies, provides valuable information on structural elements responsible for potency and selectivity.
Collapse
Affiliation(s)
- Punitha Vedantham
- Department of Chemistry, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Low WK, Dang Y, Schneider-Poetsch T, Shi Z, Choi NS, Rzasa RM, Shea HA, Li S, Park K, Ma G, Romo D, Liu JO. Isolation and identification of eukaryotic initiation factor 4A as a molecular target for the marine natural product Pateamine A. Methods Enzymol 2007; 431:303-24. [PMID: 17923240 DOI: 10.1016/s0076-6879(07)31014-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Natural products continue to demonstrate their utility both as therapeutics and as molecular probes for the discovery and mechanistic deconvolution of various cellular processes. However, this utility is dampened by the inherent difficulties involved in isolating and characterizing new bioactive natural products, in obtaining sufficient quantities of purified compound for further biological studies, and in developing bioactive probes. Key to characterizing the biological activity of natural products is the identification of the molecular target(s) within the cell. The marine sponge-derived natural product Pateamine A (PatA) has been found to be an inhibitor of eukaryotic translation initiation. Herein, we describe the methods utilized for identification of the eukaryotic translation initiation factor 4A (eIF4A) as one of the primary protein targets of PatA. We begin by describing the synthesis of an active biotin conjugate of PatA (B-PatA), made possible by total synthesis, followed by its use for affinity purification of PatA binding proteins from cellular lysates. We have attempted to present the methodology as a general technique for the identification of protein targets for small molecules including natural products.
Collapse
Affiliation(s)
- Woon-Kai Low
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Ma ZQ, Xie SX, Huang QQ, Nan FJ, Hurley TD, Ye QZ. Structural analysis of inhibition of E. coli methionine aminopeptidase: implication of loop adaptability in selective inhibition of bacterial enzymes. BMC STRUCTURAL BIOLOGY 2007; 7:84. [PMID: 18093325 PMCID: PMC2238726 DOI: 10.1186/1472-6807-7-84] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Accepted: 12/19/2007] [Indexed: 11/17/2022]
Abstract
Background Methionine aminopeptidase is a potential target of future antibacterial and anticancer drugs. Structural analysis of complexes of the enzyme with its inhibitors provides valuable information for structure-based drug design efforts. Results Five new X-ray structures of such enzyme-inhibitor complexes were obtained. Analysis of these and other three similar structures reveals the adaptability of a surface-exposed loop bearing Y62, H63, G64 and Y65 (the YHGY loop) that is an integral part of the substrate and inhibitor binding pocket. This adaptability is important for accommodating inhibitors with variations in size. When compared with the human isozymes, this loop either becomes buried in the human type I enzyme due to an N-terminal extension that covers its position or is replaced by a unique insert in the human type II enzyme. Conclusion The adaptability of the YHGY loop in E. coli methionine aminopeptidase, and likely in other bacterial methionine aminopeptidases, enables the enzyme active pocket to accommodate inhibitors of differing size. The differences in this adaptable loop between the bacterial and human methionine aminopeptidases is a structural feature that can be exploited to design inhibitors of bacterial methionine aminopeptidases as therapeutic agents with minimal inhibition of the corresponding human enzymes.
Collapse
Affiliation(s)
- Ze-Qiang Ma
- High Throughput Screening Laboratory, University of Kansas, Lawrence, Kansas 66047, USA.
| | | | | | | | | | | |
Collapse
|
141
|
Huang M, Xie SX, Ma ZQ, Huang QQ, Nan FJ, Ye QZ. Inhibition of monometalated methionine aminopeptidase: inhibitor discovery and crystallographic analysis. J Med Chem 2007; 50:5735-42. [PMID: 17948983 PMCID: PMC2522305 DOI: 10.1021/jm700930k] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Two divalent metal ions are commonly seen in the active-site cavity of methionine aminopeptidase, and at least one of the metal ions is directly involved in catalysis. Although ample structural and functional information is available for dimetalated enzyme, methionine aminopeptidase likely functions as a monometalated enzyme under physiological conditions. Information on structure, as well as catalysis and inhibition, of the monometalated enzyme is lacking. By improving conditions of high-throughput screening, we identified a unique inhibitor with specificity toward the monometalated enzyme. Kinetic characterization indicates a mutual exclusivity in binding between the inhibitor and the second metal ion at the active site. This is confirmed by X-ray structure, and this inhibitor coordinates with the first metal ion and occupies the space normally occupied by the second metal ion. Kinetic and structural analyses of the inhibition by this and other inhibitors provide insight in designing effective inhibitors of methionine aminopeptidase.
Collapse
Affiliation(s)
| | | | | | | | | | - Qi-Zhuang Ye
- * To whom correspondence should be addressed. Phone: 317 278-0304. Fax: 317 274-4686. E-Mail:
| |
Collapse
|
142
|
Klein C. Aspergillus fumigatus und Angiogenese. Von einer Zufallsentdeckung zum Rationalen Drug-Design. ACTA ACUST UNITED AC 2007; 36:450-1. [DOI: 10.1002/pauz.200700242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
143
|
Griffioen AW. Therapeutic approaches of angiogenesis inhibition: are we tackling the problem at the right level? Trends Cardiovasc Med 2007; 17:171-6. [PMID: 17574125 DOI: 10.1016/j.tcm.2007.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A growing body of evidence now demonstrates that inhibition of angiogenesis is a promising way for treatment of disease. Although the field of angiogenesis research is strongly linked to cancer biology, many other diseases were found to be dependent on angiogenesis as well, introducing a potential benefit from antiangiogenesis treatment. Recently, the first specific angiogenesis inhibitor was approved by the Food and Drug Administration for the treatment of colorectal cancer. Currently, several compounds with angiostatic activity are approved, and many are in late-stage clinical development. Most of these are indirect inhibitors, either clearing angiogenic growth factors from the circulation or blocking the signaling pathways activated by these growth factors. Although these compounds seem to represent an efficient strategy in cancer treatment, they possess an intrinsic threat to induce resistance. Therefore, it remains to be seen whether this strategy will be the most attractive in the future. Advancing insights into fundamental mechanisms will be necessary in the development of novel anticancer strategies based on inhibition of angiogenesis.
Collapse
Affiliation(s)
- Arjan W Griffioen
- Department of Pathology, Angiogenesis Laboratory, Research Institute For Growth and Development (GROW), Maastricht University, University Hospital Maastricht, The Netherlands.
| |
Collapse
|
144
|
Becker CM, D'Amato RJ. Angiogenesis and antiangiogenic therapy in endometriosis. Microvasc Res 2007; 74:121-30. [PMID: 17574280 DOI: 10.1016/j.mvr.2007.04.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Revised: 04/25/2007] [Accepted: 04/26/2007] [Indexed: 11/26/2022]
Abstract
Endometriosis, the presence of endometrium-like tissue outside of the uterine cavity, is a common disease among women of reproductive age. Typical symptoms include abdominal pain and painful menstruation. In addition, endometriosis is associated with reduced fertility. Current treatment modalities, the surgical removal of endometriotic lesions and the hormonal suppression of estrogen are associated with significant morbidity, side-effects and recurrence rates. Despite uncertainties about the pathophysiology of the disease it has recently become apparent that angiogenesis plays a pivotal role in endometriosis. This review focuses on a multitude of factors involved in the angiogenic phenotype of endometriosis demonstrating that many biological systems such as the immune system and steroid hormones are closely connected to angiogenic pathways in this disease. In addition, experimental and clinical data are discussed that concentrate on the inhibition of angiogenesis as a novel therapeutic approach for endometriosis.
Collapse
Affiliation(s)
- Christian M Becker
- Nuffield Department of Obstetrics and Gynaecology, John Radcliffe Hospital, University of Oxford, UK
| | | |
Collapse
|
145
|
Marino JP, Fisher PW, Hofmann GA, Kirkpatrick RB, Janson CA, Johnson RK, Ma C, Mattern M, Meek TD, Ryan MD, Schulz C, Smith WW, Tew DG, Tomazek TA, Veber DF, Xiong WC, Yamamoto Y, Yamashita K, Yang G, Thompson SK. Highly Potent Inhibitors of Methionine Aminopeptidase-2 Based on a 1,2,4-Triazole Pharmacophore. J Med Chem 2007; 50:3777-85. [PMID: 17636946 DOI: 10.1021/jm061182w] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
High-throughput screening for inhibitors of the human metalloprotease, methionine aminopeptidase-2 (MetAP2), identified a potent class of 3-anilino-5-benzylthio-1,2,4-triazole compounds. Efficient array and interative synthesis of triazoles led to rapid SAR development around the aniline, benzylthio, and triazole moeities. Evaluation of these analogs in a human MetAP2 enzyme assay led to the identification of several inhibitors with potencies in the 50-100 picomolar range. The deleterious effects on inhibitor potency by methylation of the anilino-triazole nitrogens, as well as the X-ray crystal structure of triazole 102 bound in the active site of MetAP2, confirm the key interactions between the triazole nitrogens, the active site cobalt atoms, and the His-231 side-chain. The structure has also provided a rationale for interpreting SAR within the triazole series. Key aniline (2-isopropylphenyl) and sulfur substituents (furanylmethyl) identified in the SAR studies led to the identification of potent inhibitors (103 and 104) of endothelial cell proliferation. Triazoles 103 and 104 also exhibited dose-dependent activity in an aortic ring tissue model of angiogenesis highlighting the potential utility of MetAP2 inhibitors as anticancer agents.
Collapse
Affiliation(s)
- Joseph P Marino
- Department of Medicinal Chemistry, Enzymology, Oncology, and Structural Biology, GlaxoSmithkline, King of Prussia, PA 19406, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Hu X, Dang Y, Tenney K, Crews P, Tsai CW, Sixt KM, Cole PA, Liu JO. Regulation of c-Src nonreceptor tyrosine kinase activity by bengamide A through inhibition of methionine aminopeptidases. CHEMISTRY & BIOLOGY 2007; 14:764-74. [PMID: 17656313 PMCID: PMC3165037 DOI: 10.1016/j.chembiol.2007.05.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 05/07/2007] [Accepted: 05/24/2007] [Indexed: 11/17/2022]
Abstract
Methionine aminopeptidases (MetAPs) remove the N-terminal initiator methionine during protein synthesis, a prerequisite step for N-terminal myristoylation. N-myristoylation of proto-oncogene c-Src is essential for its membrane association and proper signal transduction. We used bengamides, a family of general MetAP inhibitors, to understand the downstream physiological functions of MetAPs. c-Src from bengamide A-treated cells retained its N-terminal methionine and suffered a decrease in N-terminal myristoylation, which was accompanied by a shift of its subcellular distribution from the plasma membrane to the cytosol. Furthermore, bengamide A decreased the tyrosine kinase activities of c-Src both in vitro and in vivo and eventually delayed cell-cycle progression through G(2)/M. Thus, c-Src is a physiologically relevant substrate for MetAPs whose dysfunction is likely to account for the cell-cycle effects of MetAP inhibitors including bengamide A.
Collapse
Affiliation(s)
- Xiaoyi Hu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, School of Medicine, 725 N. Wolfe St. Baltimore, MD 21205
| | - Yongjun Dang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, School of Medicine, 725 N. Wolfe St. Baltimore, MD 21205
| | - Karen Tenney
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064
| | - Phillip Crews
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064
| | - Chiawei W. Tsai
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, School of Medicine, 725 N. Wolfe St. Baltimore, MD 21205
| | - Katherine M. Sixt
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University, School of Medicine, 725 N. Wolfe St. Baltimore, MD 21205
| | - Philip A. Cole
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, School of Medicine, 725 N. Wolfe St. Baltimore, MD 21205
| | - Jun O. Liu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, School of Medicine, 725 N. Wolfe St. Baltimore, MD 21205
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University, School of Medicine, 725 N. Wolfe St. Baltimore, MD 21205
| |
Collapse
|
147
|
McArdle BM, Quinn RJ. Identification of protein fold topology shared between different folds inhibited by natural products. Chembiochem 2007; 8:788-98. [PMID: 17429823 DOI: 10.1002/cbic.200700035] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Natural products have withstood the test of time as therapeutics, but new lead-generation strategies have focussed away from natural products. A new approach that uses natural-product recognition to drive an understanding of biological space might provide an impetus for renewed focus on natural-product starting points. Protein fold topology (PFT) has been shown to be an underlying factor for natural-product recognition. An investigation of natural product inhibitors of the Zincin-like fold has demonstrated their capacity also to inhibit targets of different fold types. Analysis of crystal structure complexes for natural products cocrystallised within different fold types has shown similarity at the PFT level. Two new PFT(T) (where subscript T denotes PFT shared between therapeutic targets) relationships have been established: the Zincin-like- metallohydrolase/oxidoreductase PFT(T) and the Zincin-like-phosphorylase/hydrolase PFT(T). The PFT relationship between a natural product's biosynthetic enzyme and therapeutic target, and now between different fold targets of the same natural product, suggests that PFT is the simplest descriptor of biological space. This fundamental factor for recognition could facilitate a rational approach to drug development guided by natural products.
Collapse
|
148
|
Mair B, Fuerst G, Kubitzky P, Tangl S, Bergmeister H, Losert U, Watzek G, Gruber R. The anti-angiogenic substance TNP-470 impairs peri-implant bone formation: a pilot study in the rabbit metaphysis model. Clin Oral Implants Res 2007; 18:370-5. [PMID: 17298491 DOI: 10.1111/j.1600-0501.2006.01319.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To study the osseointegration of dental implants under the conditions of impaired blood vessel formation in a rabbit model. MATERIAL AND METHODS Twenty-four titanium implants were placed in the diaphyses of 12 rabbits, two in each tibia. After 2 days, six animals received TNP-470, an anti-angiogenic substance, 10 mg/kg body weight, three times a week, by subcutaneous injection. The remaining six animals served as controls. All animals were sacrificed 6 weeks post implantation. RESULTS The amount of newly formed bone (NFB) within a peri-implant distance of 0.2 mm and the percentage of bone-to-implant contacts (BIC) were determined in undecalcified ground sections. In both groups, appositional bone growth originating from the cortex was associated with a dense network of blood vessels. Within 1 mm apical from the implant shoulder, NFB was 29.3+/-6.7% in TNP-470 treated animals vs. 44.2+/-13.1% in the controls (P=0.03). In the adjacent cortical area NFB was 46.1+/-10.0% in TNP-470 treated animals and 58.4+/-3.0% in the control group (P=0.02). In the endosteal area, 3 mm from the implant shoulder NFB was 24.8+/-11.8% in TNP-470-treated animals and 27.0+/-9.6% in controls (P=0.73) and in the most apical peri-implant area corresponding to the bone marrow cavity 4.3+/-3.2% in TNP-470-treated animals and 10.5+/-5.2% in the controls (P=0.03). No decrease in BIC in response to TNP-470 was observed. CONCLUSION The data suggest that TNP-470 decreases the formation of peri-implant bone, whereas BIC overall are not affected by it. The rabbit model presented here supports the importance of angiogenesis in the process of peri-implant bone formation.
Collapse
Affiliation(s)
- Birgit Mair
- Department of Oral Surgery, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
149
|
Bargagna-Mohan P, Hamza A, Kim YE, Ho YK(A, Mor-Vaknin N, Wendschlag N, Liu J, Evans RM, Markovitz DM, Zhan CG, Kim KB, Mohan R. The tumor inhibitor and antiangiogenic agent withaferin A targets the intermediate filament protein vimentin. CHEMISTRY & BIOLOGY 2007; 14:623-34. [PMID: 17584610 PMCID: PMC3228641 DOI: 10.1016/j.chembiol.2007.04.010] [Citation(s) in RCA: 254] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 03/30/2007] [Accepted: 04/19/2007] [Indexed: 11/22/2022]
Abstract
The natural product withaferin A (WFA) exhibits antitumor and antiangiogenesis activity in vivo, which results from this drug's potent growth inhibitory activities. Here, we show that WFA binds to the intermediate filament (IF) protein, vimentin, by covalently modifying its cysteine residue, which is present in the highly conserved alpha-helical coiled coil 2B domain. WFA induces vimentin filaments to aggregate in vitro, an activity manifested in vivo as punctate cytoplasmic aggregates that colocalize vimentin and F-actin. WFA's potent dominant-negative effect on F-actin requires vimentin expression and induces apoptosis. Finally, we show that WFA-induced inhibition of capillary growth in a mouse model of corneal neovascularization is compromised in vimentin-deficient mice. These findings identify WFA as a chemical genetic probe of IF functions, and illuminate a potential molecular target for withanolide-based therapeutics for treating angioproliferative and malignant diseases.
Collapse
Affiliation(s)
| | - Adel Hamza
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | | | - Yik Khuan (Abby) Ho
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Nirit Mor-Vaknin
- Department of Internal Medicine, Division of Infectious Diseases
| | | | - Junjun Liu
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Robert M. Evans
- Department of Pathology, University of Colorado Health Sciences Center, Denver, CO 80045, USA
| | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious Diseases
- Cellular and Molecular Biology Program, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Royce Mohan
- Department of Ophthalmology and Visual Sciences
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
150
|
Leopoldini M, Russo N, Toscano M. Which One among Zn(II), Co(II), Mn(II), and Fe(II) is the Most Efficient Ion for the Methionine Aminopeptidase Catalyzed Reaction? J Am Chem Soc 2007; 129:7776-84. [PMID: 17523636 DOI: 10.1021/ja068168t] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The catalytic hydrolysis of a methionyl-peptide substrate by a methionine aminopeptidase active site model cluster was investigated at the DF/B3LYP level of theory, in the gas-phase and in the protein environment. Zn(II), Co(II), Mn(II), and Fe(II) transition metals were examined as the potential catalytic metals of this enzyme involved in protein maturation. Two different mechanisms in which Glu204 was present as protonated or deprotonated residue were considered. The energetic profiles show lower barriers as the protonated glutamate is involved. The rate-determining step of the hydrolysis reaction is always the nucleophilic addition of the hydroxide on substrate carbon, followed by less energetically demanding methionine-peptide C-N bond scission. The lowest activation energy is obtained in the case of zinc dication while the other metals show very high energetic barriers, so that methionine aminopeptidase can be in principle recognized as a dizinc enzyme.
Collapse
Affiliation(s)
- Monica Leopoldini
- Dipartimento di Chimica and Centro di Calcolo ad Alte Prestazioni per Elaborazioni Parallele e Distribuite-Centro d'Eccellenza MIUR, Universita' della Calabria, I-87030 Arcavacata di Rende (CS), Italy
| | | | | |
Collapse
|