1
|
Sau S, Das KM, Mondal B, Thakur A. Cobalt(II)-Catalyzed Synthesis of γ-Diketones from Aryl Alkenes and Its Utilization in the Synthesis of Various Heterocyclic Compounds. J Org Chem 2024; 89:7095-7108. [PMID: 38701377 DOI: 10.1021/acs.joc.4c00487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
An earth-abundant Co(II) salt-catalyzed mild and affordable synthetic route has been developed for the synthesis of industrially relevant 1,4-dicarbonyl compounds (or γ-diketones) via oxidative coupling between aryl alkenes and ketones (both cyclic and acyclic) using TBHP and DBU as the oxidant and base, respectively. 1,4-Dicarbonyl compounds are known to be synthesized using expensive metal catalysts, dual catalysts, or low-cost metal complexes combined with an additive or ligand template, which further needs to be synthesized. Herein, we report the synthesis of 1,4-dicarbonyl compounds using cobalt(II) acetate as a catalyst without any expensive co-catalyst or ligand templates. This methodology has a broad substrate scope with significant yields and good functional group tolerance. Generation of unsymmetrical 1,4-dicarbonyls at room temperature and its versatile synthetic expansion to produce synthetically and biologically valuable heterocyclic compounds are salient features of this novel methodology. In addition, various controlled experiments such as primary kinetic isotope effect study, Hammett analysis with variation of the nature of the substituents on the styrene ring, and theoretical calculations (density functional theory) unravel the mechanistic intricacies involved in this new, simple, and atom-economic methodology.
Collapse
Affiliation(s)
- Subham Sau
- Department of Chemistry, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Krishna Mohan Das
- Department of Chemistry, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Bijan Mondal
- Institut für Anorganische Chemie, Universität Regensburg, Universität Strasse 31, 93040 Regensburg, Germany
| | - Arunabha Thakur
- Department of Chemistry, Jadavpur University, Kolkata 700032, West Bengal, India
| |
Collapse
|
2
|
Palani P, Arumugam A, Raja D, Muthu K, Senadi GC. Photoredox-catalyzed 1,2-oxo-alkylation of vinyl arenes with 1,3-diketones: an approach to 1,4-dicarbonyls via C-C activation. Chem Commun (Camb) 2023; 59:11433-11436. [PMID: 37671608 DOI: 10.1039/d3cc02366d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
A mild and inexpensive approach to synthesising a series of 1,4-diketones in moderate to excellent yields via 1,2-oxo alkylation has been developed using fluorescein as a photocatalyst and air as an oxidant. The key features include (i) varied substrate scope (39 examples); (ii) good functional group tolerance; (iii) unsymmetrical 1,4-dicarbonyls; (iv) late-stage functionalization of thymol and ibuprofen derivatives; and (v) the synthetic expansion to 5- and 6-membered N-, O- and S-containing heterocycles.
Collapse
Affiliation(s)
- Pushbaraj Palani
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur-603 203, Chengalpattu District, Tamil Nadu, India.
| | - Ajithkumar Arumugam
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur-603 203, Chengalpattu District, Tamil Nadu, India.
| | - Dineshkumar Raja
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur-603 203, Chengalpattu District, Tamil Nadu, India.
| | - Kesavan Muthu
- Interdisciplinary Institute of Indian System of Medicine (IIISM), SRM Institute of Science and Technology, SRM Nagar, Kattankulathur-603 203, Chengalpattu District, Tamil Nadu, India
| | - Gopal Chandru Senadi
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur-603 203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
3
|
Prado AF, Batista RIM, Tanus-Santos JE, Gerlach RF. Matrix Metalloproteinases and Arterial Hypertension: Role of Oxidative Stress and Nitric Oxide in Vascular Functional and Structural Alterations. Biomolecules 2021; 11:biom11040585. [PMID: 33923477 PMCID: PMC8074048 DOI: 10.3390/biom11040585] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Various pathophysiological mechanisms have been implicated in hypertension, but those resulting in vascular dysfunction and remodeling are critical and may help to identify critical pharmacological targets. This mini-review article focuses on central mechanisms contributing to the vascular dysfunction and remodeling of hypertension, increased oxidative stress and impaired nitric oxide (NO) bioavailability, which enhance vascular matrix metalloproteinase (MMP) activity. The relationship between NO, MMP and oxidative stress culminating in the vascular alterations of hypertension is examined. While the alterations of hypertension are not fully attributable to these pathophysiological mechanisms, there is strong evidence that such mechanisms play critical roles in increasing vascular MMP expression and activity, thus resulting in abnormal degradation of extracellular matrix components, receptors, peptides, and intracellular proteins involved in the regulation of vascular function and structure. Imbalanced vascular MMP activity promotes vasoconstriction and impairs vasodilation, stimulating vascular smooth muscle cells (VSMC) to switch from contractile to synthetic phenotypes, thus facilitating cell growth or migration, which is associated with the deposition of extracellular matrix components. Finally, the protective effects of MMP inhibitors, antioxidants and drugs that enhance vascular NO activity are briefly discussed. Newly emerging therapies that address these essential mechanisms may offer significant advantages to prevent vascular remodeling in hypertensive patients.
Collapse
Affiliation(s)
- Alejandro F. Prado
- Laboratory of Structural Biology, Institute of Biological Sciences, Federal University of Para, Belem, PA 66075-110, Brazil;
| | - Rose I. M. Batista
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; (R.I.M.B.); (J.E.T.-S.)
| | - Jose E. Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; (R.I.M.B.); (J.E.T.-S.)
| | - Raquel F. Gerlach
- Department of Morphology, Physiology and Basic Pathology, Faculty of Dentistry of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP 14040-904, Brazil
- Correspondence: ; Tel.: +55-16-33154065
| |
Collapse
|
4
|
CMT-308, a Nonantimicrobial Chemically-Modified Tetracycline, Exhibits Anti-Melanogenic Activity by Suppression of Melanosome Export. Biomedicines 2020; 8:biomedicines8100411. [PMID: 33066033 PMCID: PMC7601524 DOI: 10.3390/biomedicines8100411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/15/2022] Open
Abstract
CMT-308 is a nonantimicrobial chemically-modified tetracycline (CMT), which we have previously shown exhibits antifungal activity and pleiotropic anti-inflammatory activities, including inhibition of the enzymatic activity of matrix metalloproteinases (MMPs). Based on its chemical structure, we hypothesized that CMT-308 could inhibit melanogenesis and might be a candidate for the treatment of skin hyperpigmentation disorders which occur due to unregulated melanin biosynthesis and/or transport. CMT-308 was first studied for any effects on activity of the enzyme tyrosinase in vitro using a purified preparation of mushroom tyrosinase; the mode of inhibition of the soluble fungal enzyme was evaluated by Lineweaver-Burk and Dixon plots as well as by non-linear least squares fitting. Next, the effects of CMT-308 were tested in mammalian cell cultures using B16F10 mouse melanoma cells and further validated in darkly-pigmented human melanocytes (HEMn-DP). Our results showed that micromolar concentrations of CMT-308 inhibited mushroom tyrosinase enzyme activity, using the first two substrates in the melanogenesis pathway (l-tyrosine and l-3,4-dihydroxyphenylalanine (l-DOPA)); CMT-308 inhibited mushroom tyrosinase primarily via a mixed mode of inhibition, with the major contribution from a competitive mode. In B16F10 cell cultures, CMT-308 (10 µM) significantly diminished total melanin levels with a selective reduction of extracellular melanin levels, under both basal and hormone-stimulated conditions without any cytotoxicity over a duration of 72 h. Studies of potential mechanisms of inhibition of melanogenesis in B16F10 cells showed that, in mammalian cells, CMT-308 did not inhibit intracellular tyrosinase activity or the activity of α-glucosidase, an enzyme that regulates maturation of tyrosinase. However, CMT-308 suppressed MITF protein expression in B16F10 cells and showed copper chelating activity and antioxidant activity in a cell-free system. The significantly lower extracellular melanin levels obtained at 10 µM indicate that CMT-308’s anti-melanogenic action may be attributed to a selective inhibition of melanosome export with the perinuclear aggregation of melanosomes, rather than a direct effect on the tyrosinase-catalyzed steps in melanin biosynthesis. These results were validated in HEMn-DP cells where CMT-308 suppressed dendricity in a fully reversible manner without affecting intracellular melanin synthesis. Furthermore, the capacity of CMT-308 to inhibit melanosome export was retained in cocultures of HEMn-DP and HaCaT. In summary, our results offer promise for therapeutic strategies to combat the effects of hyperpigmentation by use of CMT-308 at low micromolar concentrations.
Collapse
|
5
|
Raeeszadeh-Sarmazdeh M, Do LD, Hritz BG. Metalloproteinases and Their Inhibitors: Potential for the Development of New Therapeutics. Cells 2020; 9:E1313. [PMID: 32466129 PMCID: PMC7290391 DOI: 10.3390/cells9051313] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023] Open
Abstract
The metalloproteinase (MP) family of zinc-dependent proteases, including matrix metalloproteinases (MMPs), a disintegrin and metalloproteases (ADAMs), and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs) plays a crucial role in the extracellular matrix (ECM) remodeling and degradation activities. A wide range of substrates of the MP family includes ECM components, chemokines, cell receptors, and growth factors. Metalloproteinases activities are tightly regulated by proteolytic activation and inhibition via their natural inhibitors, tissue inhibitors of metalloproteinases (TIMPs), and the imbalance of the activation and inhibition is responsible in progression or inhibition of several diseases, e.g., cancer, neurological disorders, and cardiovascular diseases. We provide an overview of the structure, function, and the multifaceted role of MMPs, ADAMs, and TIMPs in several diseases via their cellular functions such as proteolysis of other cell signaling factors, degradation and remodeling of the ECM, and other essential protease-independent interactions in the ECM. The significance of MP inhibitors targeting specific MMP or ADAMs with high selectivity is also discussed. Recent advances and techniques used in developing novel MP inhibitors and MP responsive drug delivery tools are also reviewed.
Collapse
Affiliation(s)
- Maryam Raeeszadeh-Sarmazdeh
- Chemical and Materials Engineering Department, University of Nevada, Reno, NV 89557, USA; (L.D.D.); (B.G.H.)
| | | | | |
Collapse
|
6
|
Young CJ, Richard K, Beruar A, Lo SY, Siemann S. An investigation of the pH dependence of copper-substituted anthrax lethal factor and its mechanistic implications. J Inorg Biochem 2018; 182:1-8. [PMID: 29407865 DOI: 10.1016/j.jinorgbio.2018.01.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/18/2017] [Accepted: 01/22/2018] [Indexed: 10/18/2022]
Abstract
Anthrax lethal factor (LF) is a zinc-dependent endopeptidase involved in the cleavage of proteins critical to the maintenance of host signaling pathways during anthrax infections. Although zinc is typically regarded as the native metal ion in vivo, LF is highly tolerant to metal substitution, with its replacement by copper yielding an enzyme (CuLF) 4.5-fold more active than the native zinc protein (at pH 7). The current study demonstrates that by careful choice of the buffer, ionic strength, pH and substrate, the activity ratio of CuLF and native LF can be increased to >40-fold. Using a fluorogenic LF substrate, such optimized assay conditions can be exploited to detect LF concentrations as low as 2 pM. In contrast to the zinc form, CuLF was found to be inhibited by bromide and iodide ions, to be resistant to metal loss under acidic conditions, and to display a sharp pH dependence with significantly shifted alkaline limb towards more acidic conditions. The alkaline limb in the enzyme's pH profile is suggested to originate from changes in the protonation state of the metal-bound water molecule which serves as the nucleophile in the catalytic mechanism. Based on these observations and studies on other zinc proteases, a minimal mechanism for LF is proposed.
Collapse
Affiliation(s)
- Calvin J Young
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Kaitlin Richard
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Ananya Beruar
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Suet Y Lo
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Stefan Siemann
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada.
| |
Collapse
|
7
|
Ni Y, Tang Z, Yang J, Gao Y, Lin H, Guo L, Zhang K, Zhang X. Collagen structure regulates MSCs behavior by MMPs involved cell–matrix interactions. J Mater Chem B 2018; 6:312-326. [DOI: 10.1039/c7tb02377d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Various scaffolds have been studied in the formation of cell niches and regulation of mesenchymal stem cells (MSCs) behaviors.
Collapse
Affiliation(s)
- Yilu Ni
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Zhurong Tang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Jirong Yang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yongli Gao
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Hai Lin
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Likun Guo
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Kai Zhang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| |
Collapse
|
8
|
Li X, Ma D, Zha X, Quan D, Pan D, Sun M, Hu B, Zhao B. Ilomastat, a synthetic inhibitor of MMPs, prevents lung injury induced by γ-ray irradiation in mice. Oncotarget 2017; 8:60789-60808. [PMID: 28977826 PMCID: PMC5617386 DOI: 10.18632/oncotarget.18487] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/05/2017] [Indexed: 01/27/2023] Open
Abstract
Lung injury is one of the pathological features in human or animal after radiation and the main side effect for patient after lung cancer radiotherapy. The efficient protective strategy still needs to exploit and the underlying mechanisms remain to be investigated. We found that the expression and activity of matrix metalloproteinases (MMPs) significantly increased at the early stage of radiation-induced lung injury (RILI). Pretreatment with Ilomastat, a synthetic inhibitor of MMPs, decreased the expression and activity of MMPs and significantly alleviated the lung inflammation and fibrosis in the irradiated mice, as well as enhanced the survival of irradiated mice. In addition, the levels of TGF-β, IL-6, TNF-α and IL-1β in the tissues dramatically reduced in the irradiated mice pretreated with Ilomastat. Furthermore, our experiments in vitro also showed that radiation significantly increased the MMPs activity, and Ilomastat pretreatment inhibited the activity of MMPs activated by irradiation and increased the cell survival. It is the first report, to our knowledge, to demonstrate that Ilomastat is a potential effective reliever for RILI and MMPs may play important roles in the process of RILI.
Collapse
Affiliation(s)
- Xiaoman Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China.,CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Space Radiobiology of Gansu Province, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Dehui Ma
- College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tong Liao, China
| | - Xiaodan Zha
- College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tong Liao, China
| | - Dongqin Quan
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | - Dong Pan
- CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Space Radiobiology of Gansu Province, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Manji Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | - Burong Hu
- CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Space Radiobiology of Gansu Province, Lanzhou, China
| | - Baoquan Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Amin M, Pushpakumar S, Muradashvili N, Kundu S, Tyagi SC, Sen U. Regulation and involvement of matrix metalloproteinases in vascular diseases. FRONT BIOSCI-LANDMRK 2016; 21:89-118. [PMID: 26709763 PMCID: PMC5462461 DOI: 10.2741/4378] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc dependent endopeptidases whose main function is to degrade and deposit structural proteins within the extracellular matrix (ECM). A dysregulation of MMPs is linked to vascular diseases. MMPs are classified into collagenases, gelatinases, membrane-type, metalloelastase, stromelysins, matrilysins, enamelysins, and unclassified subgroups. The production of MMPs is stimulated by factors such as oxidative stress, growth factors and inflammation which lead to its up- or down-regulation with subsequent ECM remodeling. Normally, excess activation of MMPs is controlled by their endogenous inhibitors, tissue inhibitors of metalloproteinases (TIMPs). An imbalance of MMPs and TIMPs has been implicated in hypertension, atherosclerotic plaque formation and instability, aortic aneurysms and varicose vein wall remodeling. Also, recent evidence suggests epigenetic regulation of some MMPs in angiogenesis and atherosclerosis. Over the years, pharmacological inhibitors of MMPs have been used to modify or prevent the development of the disease with some success. In this review, we discuss recent advances in MMP biology, and their involvement in the manifestation of vascular disease.
Collapse
Affiliation(s)
- Matthew Amin
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Sathnur Pushpakumar
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Sourav Kundu
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Utpal Sen
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202,
| |
Collapse
|
10
|
Ambrose EA. Botulinum Neurotoxin, Tetanus Toxin, and Anthrax Lethal Factor Countermeasures. TOPICS IN MEDICINAL CHEMISTRY 2016. [DOI: 10.1007/7355_2016_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Kurbanov EK, Chiu TL, Solberg J, Francis S, Maize KM, Fernandez J, Johnson RL, Hawkinson JE, Walters MA, Finzel BC, Amin EA. Probing the S2′ Subsite of the Anthrax Toxin Lethal Factor Using Novel N-Alkylated Hydroxamates. J Med Chem 2015; 58:8723-33. [DOI: 10.1021/acs.jmedchem.5b01446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Elbek K. Kurbanov
- Department
of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Ting-Lan Chiu
- Department
of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Jonathan Solberg
- Institute
for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Subhashree Francis
- Institute
for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Kimberly M. Maize
- Department
of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Jenna Fernandez
- Department
of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Rodney L. Johnson
- Department
of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Jon E. Hawkinson
- Institute
for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Michael A. Walters
- Institute
for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Barry C. Finzel
- Department
of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Elizabeth Ambrose Amin
- Department
of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55414, United States
- Minnesota
Supercomputing Institute for Advanced Computational Research, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
12
|
Bouzianas DG. Potential biological targets ofBacillus anthracisin anti-infective approaches against the threat of bioterrorism. Expert Rev Anti Infect Ther 2014; 5:665-84. [PMID: 17678429 DOI: 10.1586/14787210.5.4.665] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The terrorist attacks of 2001 involving anthrax underscore the imperative that safe and effective medical countermeasures should be readily available. Vaccination appears to be the most effective form of mass protection against a biological attack, but the current vaccines have drawbacks that justify the enormous amount of effort currently being put into developing more effective vaccines and other treatment modalities. After providing a comprehensive overview of the organism Bacillus anthracis as a biological weapon and its pathogenicity, this review briefly summarizes the current knowledge vital to the management of anthrax disease. This knowledge has been acquired since 2001 as a result of the progress on anthrax research and focuses on the possible development of improved human anti-infective strategies targeting B. anthracis spore components, as well as strategies based on host-pathogen interactions.
Collapse
Affiliation(s)
- Dimitrios G Bouzianas
- Department of Medical Laboratories, Faculty of Health and Care Professions, University-level Technological Educational Institute of Thessaloniki, Greece.
| |
Collapse
|
13
|
Antonelli AC, Zhang Y, Golub LM, Johnson F, Simon SR. Inhibition of anthrax lethal factor by curcumin and chemically modified curcumin derivatives. J Enzyme Inhib Med Chem 2013; 29:663-9. [PMID: 24102525 PMCID: PMC4196590 DOI: 10.3109/14756366.2013.837901] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Curcuma longa Curcumin (diferuloylmethane), the active ingredient in the eastern spice turmeric (Curcuma longa), has been shown to inhibit the activities of numerous enzymes and signaling molecules involved in cancer, bacterial and viral infections and inflammatory diseases. We have investigated the inhibitory activities of curcumin and chemically modified curcumin (CMC) derivatives toward lethal factor (LF), the proteolytic component of anthrax toxin produced by the bacterium Bacillus anthracis. Curcumin (Compound 1) appears to inhibit the catalytic activity of LF through a mixture of inhibitory mechanisms, without significant compromise to the binding of oligopeptide substrates, and one CMC derivative in particular, Compound 3 (4-phenylaminocarbonylbis-demethoxycurcumin), is capable of inhibiting LF with potency comparable with the parent compound, while also showing improved solubility and stability. The quantitative reduction in catalytic activity achieved by the different CMC derivatives appears to be a function of the proportion of the multiple mechanisms through which they inhibit the enzyme.
Collapse
|
14
|
Chiu TL, Amin EA. Development of a comprehensive, validated pharmacophore hypothesis for anthrax toxin lethal factor (LF) inhibitors using genetic algorithms, Pareto scoring, and structural biology. J Chem Inf Model 2012; 52:1886-97. [PMID: 22697455 PMCID: PMC3477282 DOI: 10.1021/ci300121p] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Anthrax is an acute infectious disease caused by the spore-forming bacterium Bacillus anthracis. The anthrax toxin lethal factor (LF), an 89-kDa zinc hydrolase secreted by the bacilli, is the toxin component chiefly responsible for pathogenesis and has been a popular target for rational and structure-based drug design. Although hundreds of small-molecule compounds have been designed to target the LF active site, relatively few reported inhibitors have exhibited activity in cell-based assays, and no LF inhibitor is currently available to treat or prevent anthrax. This study presents a new pharmacophore map assembly, validated by experiment, designed to rapidly identify and prioritize promising LF inhibitor scaffolds from virtual compound libraries. The new hypothesis incorporates structural information from all five available LF enzyme-inhibitor complexes deposited in the Protein Data Bank (PDB) and is the first LF pharmacophore map reported to date that includes features representing interactions involving all three key subsites of the LF catalytic binding region. In a wide-ranging validation study on all 546 compounds for which published LF biological activity data exist, this model displayed strong selectivity toward nanomolar-level LF inhibitors, successfully identifying 72.1% of existing nanomolar-level compounds in an unbiased test set, while rejecting 100% of weakly active (>100 μM) compounds. In addition to its capabilities as a database searching tool, this comprehensive model points to a number of key design principles and previously unidentified ligand-receptor interactions that are likely to influence compound potency.
Collapse
Affiliation(s)
- Ting-Lan Chiu
- Department of Medicinal Chemistry and Minnesota Supercomputing Institute for Advanced Computational Research, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Elizabeth A. Amin
- Department of Medicinal Chemistry and Minnesota Supercomputing Institute for Advanced Computational Research, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| |
Collapse
|
15
|
Artenstein AW, Opal SM. Novel approaches to the treatment of systemic anthrax. Clin Infect Dis 2012; 54:1148-61. [PMID: 22438345 DOI: 10.1093/cid/cis017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Anthrax continues to generate concern as an agent of bioterrorism and as a natural cause of sporadic disease outbreaks. Despite the use of appropriate antimicrobial agents and advanced supportive care, the mortality associated with the systemic disease remains high. This is primarily due to the pathogenic exotoxins produced by Bacillus anthracis as well as other virulence factors of the organism. For this reason, new therapeutic strategies that target events in the pathogenesis of anthrax and may potentially augment antimicrobials are being investigated. These include anti-toxin approaches, such as passive immune-based therapies; non-antimicrobial drugs with activity against anthrax toxin components; and agents that inhibit binding, processing, or assembly of toxins. Adjunct therapies that target spore germination or downstream events in anthrax intoxication are also under investigation. In combination, these modalities may enhance the management of systemic anthrax.
Collapse
Affiliation(s)
- Andrew W Artenstein
- Center for Biodefense and Emerging Pathogens, Department of Medicine, Memorial Hospital of Rhode Island, Pawtucket, and The Warren Alpert Medical School of Brown University, Providence, RI 02860, USA
| | | |
Collapse
|
16
|
Koçer SS, Wang HY, Malbon CC. "Shaping" of cell signaling via AKAP-tethered PDE4D: Probing with AKAR2-AKAP5 biosensor. J Mol Signal 2012; 7:4. [PMID: 22583680 PMCID: PMC3493269 DOI: 10.1186/1750-2187-7-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 04/15/2012] [Indexed: 11/25/2022] Open
Abstract
Background PKA, a key regulator of cell signaling, phosphorylates a diverse and important array of target molecules and is spatially docked to members of the A-kinase Anchoring Protein (AKAP) family. AKAR2 is a biosensor which yields a FRET signal in vivo, when phosphorylated by PKA. AKAP5, a prominent member of the AKAP family, docks several signaling molecules including PKA, PDE4D, as well as GPCRs, and is obligate for the propagation of the activation of the mitogen-activated protein kinase cascade from GPCRs to ERK1,2. Results Using an AKAR2-AKAP5 fusion “biosensor”, we investigated the spatial-temporal activation of AKAP5 undergoing phosphorylation by PKA in response to β-adrenergic stimulation. The pattern of PKA activation reported by AKAR2-AKAP5 is a more rapid and spatially distinct from those “sensed” by AKAR2-AKAP12. Spatial-temporal restriction of activated PKA by AKAP5 was found to “shape” the signaling response. Phosphatase PDE4D tethered to AKAP5 also later reverses within 60 s elevated intracellular cyclic AMP levels stimulated by β-adrenergic agonist. AKAP12, however, fails to attenuate the rise in cyclic AMP over this time. Fusion of the AKAP5 PDE4D-binding-domain to AKAP12 was found to accelerate a reversal of accumulation of intracellular cyclic AMP. Conclusion AKAPs, which are scaffolds with tethered enzymes, can “shape” the temporal and spatial aspects of cell signaling.
Collapse
Affiliation(s)
- Salih S Koçer
- Department of Pharmacological Sciences, Health Sciences Center, BST-7, SUNY at Stony Brook, School of Medicine, Stony Brook, New York 11794-8651, USA.
| | | | | |
Collapse
|
17
|
Abstract
The MEROPS website (http://merops.sanger.ac.uk) includes information on peptidase inhibitors as well as on peptidases and their substrates. Displays have been put in place to link peptidases and inhibitors together. The classification of protein peptidase inhibitors is continually being revised, and currently inhibitors are grouped into 67 families based on comparisons of protein sequences. These families can be further grouped into 38 clans based on comparisons of tertiary structure. Small molecule inhibitors are important reagents for peptidase characterization and, with the increasing importance of peptidases as drug targets, they are also important to the pharmaceutical industry. Small molecule inhibitors are now included in MEROPS and over 160 summaries have been written.
Collapse
Affiliation(s)
- Neil D Rawlings
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK.
| |
Collapse
|
18
|
Chiu TL, Solberg J, Patil S, Geders TW, Zhang X, Rangarajan S, Francis R, Finzel BC, Walters MA, Hook DJ, Amin EA. Identification of novel non-hydroxamate anthrax toxin lethal factor inhibitors by topomeric searching, docking and scoring, and in vitro screening. J Chem Inf Model 2009; 49:2726-34. [PMID: 19928768 PMCID: PMC2805240 DOI: 10.1021/ci900186w] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Anthrax is an infectious disease caused by Bacillus anthracis, a Gram-positive, rod-shaped, anaerobic bacterium. The lethal factor (LF) enzyme is secreted by B. anthracis as part of a tripartite exotoxin and is chiefly responsible for anthrax-related cytotoxicity. As LF can remain in the system long after antibiotics have eradicated B. anthracis from the body, the preferred therapeutic modality would be the administration of antibiotics together with an effective LF inhibitor. Although LF has garnered a great deal of attention as an attractive target for rational drug design, relatively few published inhibitors have demonstrated activity in cell-based assays and, to date, no LF inhibitor is available as a therapeutic or preventive agent. Here we present a novel in silico high-throughput virtual screening protocol that successfully identified 5 non-hydroxamic acid small molecules as new, preliminary LF inhibitor scaffolds with low micromolar inhibition against that target, resulting in a 12.8% experimental hit rate. This protocol screened approximately 35 million nonredundant compounds for potential activity against LF and comprised topomeric searching, docking and scoring, and drug-like filtering. Among these 5 hit compounds, none of which has previously been identified as a LF inhibitor, three exhibited experimental IC(50) values less than 100 microM. These three preliminary hits may potentially serve as scaffolds for lead optimization as well as templates for probe compounds to be used in mechanistic studies. Notably, our docking simulations predicted that these novel hits are likely to engage in critical ligand-receptor interactions with nearby residues in at least two of the three (S1', S1-S2, and S2') subsites in the LF substrate binding area. Further experimental characterization of these compounds is in process. We found that micromolar-level LF inhibition can be attained by compounds with non-hydroxamate zinc-binding groups that exhibit monodentate zinc chelation as long as key hydrophobic interactions with at least two LF subsites are retained.
Collapse
Affiliation(s)
- Ting-Lan Chiu
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Jonathan Solberg
- Institute for Therapeutics Discovery and Development, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Satish Patil
- Department of Chemistry, University of Minnesota, 207 Pleasant St. SE, Minneapolis, MN 55455-0431
| | - Todd W. Geders
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Xia Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Subhashree Rangarajan
- Institute for Therapeutics Discovery and Development, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Rawle Francis
- Institute for Therapeutics Discovery and Development, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Barry C. Finzel
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Michael A. Walters
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
- Institute for Therapeutics Discovery and Development, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Derek J. Hook
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
- Institute for Therapeutics Discovery and Development, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Elizabeth A. Amin
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
- Minnesota Supercomputing Institute for Advanced Computational Research, 117 Pleasant St. SE, Minneapolis, MN 55455
| |
Collapse
|
19
|
Koçer SS, Matic M, Ingrassia M, Walker SG, Roemer E, Licul G, Simon SR. Effects of anthrax lethal toxin on human primary keratinocytes. J Appl Microbiol 2009; 105:1756-67. [PMID: 19120626 DOI: 10.1111/j.1365-2672.2008.03806.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS To investigate the effects of anthrax lethal toxin (LeTx) on human primary keratinocytes. METHODS AND RESULTS We show here that human primary keratinocytes are resistant to LeTx-triggered cytotoxicity. All but one of the MEKs (mitogen-activated protein kinase kinases) are cleaved within 3 h, and the cleavage of MEKs in keratinocytes leads to their subsequent proteasome-mediated degradation at different rates. Moreover, LeTx reduced the concentration of several cytokines except RANTES in culture. CONCLUSIONS Our results indicate that primary keratinocytes are resistant to LeTx cytotoxicity, and MEK cleavage does not correlate with LeTx cytotoxicity. Although LeTx is considered as an anti-inflammatory agent, it upregulates RANTES. SIGNIFICANCE AND IMPACT OF THE STUDY According to a current view, the action of LeTx results in downregulation of the inflammatory response, as evidenced by diminished expression of several inflammatory biomarkers. Paradoxically, LeTx has been reported to attract neutrophils to cutaneous infection sites. This paper, which shows that RANTES, a chemoattractant for immune cells, is upregulated after exposure of keratinocytes to LeTx, although a number of other markers of the inflammatory response are downregulated. Our results might explain why the exposure of keratinocytes to LeTx results in the recruitment of neutrophils to cutaneous infection sites, while the expression of several inflammatory biomarkers is diminished.
Collapse
Affiliation(s)
- S S Koçer
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, New York, NY 11794-8691, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Tonello F, Montecucco C. The anthrax lethal factor and its MAPK kinase-specific metalloprotease activity. Mol Aspects Med 2009; 30:431-8. [PMID: 19665472 DOI: 10.1016/j.mam.2009.07.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 07/30/2009] [Indexed: 02/06/2023]
Abstract
The anthrax lethal factor is a multi-domain protein toxin released by Bacillus anthracis which enters cells in a process mediated by the protective antigen and specific cell receptors. In the cytosol, the lethal factor cleaves the N-terminal tail of many MAPK kinases, thus deranging a major cell signaling pathway. The structural features at the basis of these activities of LF are reviewed here with particular attention to the proteolytic activity and to the identification of specific inhibitors. A significant similarity between the metalloprotease domain of the lethal factor and of that of the clostridial neurotoxins has been noted and is discussed.
Collapse
Affiliation(s)
- Fiorella Tonello
- Dipartimento di Scienze Biomediche Sperimentali, Istituto CNR di Neuroscienze, Università di Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | | |
Collapse
|
21
|
Johnson SL, Chen LH, Barille E, Emdadi A, Sabet M, Yuan H, Wei J, Guiney D, Pellecchia M. Structure-activity relationship studies of a novel series of anthrax lethal factor inhibitors. Bioorg Med Chem 2009; 17:3352-68. [PMID: 19359184 PMCID: PMC2730741 DOI: 10.1016/j.bmc.2009.03.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Revised: 03/13/2009] [Accepted: 03/20/2009] [Indexed: 10/21/2022]
Abstract
We report on the identification of a novel small molecule inhibitor of anthrax lethal factor using a high-throughput screening approach. Guided by molecular docking studies, we carried out structure-activity relationship (SAR) studies and evaluated activity and selectivity of most promising compounds in in vitro enzyme inhibition assays and cellular assays. Selected compounds were further analyzed for their in vitro ADME properties, which allowed us to select two compounds for further preliminary in vivo efficacy studies. The data provided represents the basis for further pharmacology and medicinal chemistry optimizations that could result in novel anti-anthrax therapies.
Collapse
Affiliation(s)
- Sherida L. Johnson
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Li-Hsing Chen
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Elisa Barille
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Aras Emdadi
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Mojgan Sabet
- Department of Medicine, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Hongbin Yuan
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Jun Wei
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Donald Guiney
- Department of Medicine, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Maurizio Pellecchia
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| |
Collapse
|
22
|
Gratacós-Cubarsí M, Castellari M, Hortós M, García-Regueiro JA, Lametsch R, Jessen F. Effects of tetracycline administration on the proteomic profile of pig muscle samples (L. dorsi). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2008; 56:9312-9316. [PMID: 18778074 DOI: 10.1021/jf801390z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Effect of tetracycline (TC) administration on the proteomic profile of pig muscle was evaluated by 2D electrophoresis and MALDI-TOF mass spectrometry. The TC content at slaughter was determined in L. dorsi samples by HPLC-DAD. Mean residual concentration of TC in the muscle of treated animals, calculated as the sum of TC and epi-TC was 126.3 microg/kg, indicating a rapid elimination of TC in this tissue. Several differential spots (n = 54, p < 0.05) were observed in protein profiles from control and treated animals. MALDI-TOF identification gave a positive match for 5 differential spots, that is, glycerol-3-phosphate dehydrogenase 1 (G3PD1), phosphoglycerate kinase 1, novelprotein (0610037L13Rik), leucine aminopeptidase 3 (LAP), and hypothetical protein isoform 2. Results show that proteomics could be a useful tool to reveal pharmacological treatments with TC, even if the possible uses of differential spots as biomarkers to detect illegal administration of TC require further studies. Different spot patterns as a consequence of TC treatments seem to be another interesting issue for the consequences on tissue metabolism and meat quality.
Collapse
Affiliation(s)
- M Gratacós-Cubarsí
- Food Chemistry Unit, IRTA-Monells, Finca Camps i Armet s/n, Monells, Girona, Spain.
| | | | | | | | | | | |
Collapse
|
23
|
Knockenhauer KE, Sawicka KM, Roemer EJ, Simon SR. Protective antigen composite nanofibers as a transdermal anthrax vaccine. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2008; 2008:1040-1043. [PMID: 19162840 DOI: 10.1109/iembs.2008.4649337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Anthrax, a disease caused by the gram positive bacteria Bacillus anthracis, has become an increasing threat to public health in the last several years, due to its use as an agent of biological warfare. The currently utilized human anthrax vaccine, which confers immunity through the host antibody recognition of protective antigen (PA), requires a three dose regimen and annual booster shots after the initial vaccination to maintain its efficacy. The long term goal of this project is to produce an anthrax vaccine that is capable of delivering protective antigen through human skin. The novel method for transdermal vaccine delivery that we propose utilizes the high surface area to volume ratio offered by protein-containing nanofiber membranes, prepared by the electrospinning technique. Research has already been undertaken to study the effect the main virulent agent of anthrax, lethal toxin (LT), has on a human monocytic cell line, Monomac 6 cells (MM6). Lethal toxin is said to comprise of a Zn2+ -dependent metalloprotease known as lethal factor (LF), and a binding protein known as protective antigen. The successful encapsulation of the protective antigen within the nanofibrous membrane was analyzed with the use of an in vitro MM6 assay. The assay was designed to ensure the functionality of PA through the harsh environment of the electrospinning process. Quantitative analysis of IL-6 cytokine production by lipopolysaccharide (LPS) stimulated MM6 cells in the presence of LF and PA provided proof that PA retained its biological activity through the process of electrospinning. This finding provides an innovative platform for the development of a transdermal anthrax vaccine.
Collapse
|
24
|
Schepetkin IA, Khlebnikov AI, Kirpotina LN, Quinn MT. Novel small-molecule inhibitors of anthrax lethal factor identified by high-throughput screening. J Med Chem 2006; 49:5232-44. [PMID: 16913712 DOI: 10.1021/jm0605132] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Anthrax lethal factor (LF) is a key virulence factor of anthrax lethal toxin. We screened a chemolibrary of 10,000 drug-like molecules for their ability to inhibit LF and identified 18 novel small molecules with potent LF inhibitory activity. Three additional LF inhibitors were identified through further structure-activity relationship (SAR) analysis. All 21 compounds inhibited LF with an IC50 range of 0.8 to 11 muM, utilizing mixed-mode competitive inhibition. An evaluation of inhibitory activity against a range of unrelated proteases showed relatively high specificity for LF. Furthermore, pharmacophore modeling of these compounds showed a high degree of similarity to the model published by Panchal et al. (Nat. Struct. Mol. Biol. 2004, 11, 67-72), indicating that the conformational features of these inhibitors are structurally compatible with the steric constraints of the substrate-binding pocket. These novel LF inhibitors and the structural scaffolds identified as important for inhibitory activity represent promising leads to pursue for further LF inhibitor development.
Collapse
Affiliation(s)
- Igor A Schepetkin
- Department of Veterinary Molecular Biology, Montana State University, Bozeman, Montana 59717, USA
| | | | | | | |
Collapse
|
25
|
Jiao GS, Simo O, Nagata M, O'Malley S, Hemscheidt T, Cregar L, Millis SZ, Goldman ME, Tang C. Selectively guanidinylated derivatives of neamine. Syntheses and inhibition of anthrax lethal factor protease. Bioorg Med Chem Lett 2006; 16:5183-9. [PMID: 16870442 DOI: 10.1016/j.bmcl.2006.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Revised: 06/26/2006] [Accepted: 07/05/2006] [Indexed: 11/25/2022]
Abstract
A series of mono-, di-, and tri-guanidinylated derivatives of neamine were prepared via selective guanidinylation of neamine. These molecules represent a novel scaffold as inhibitors of anthrax lethal factor zinc metalloprotease. Methods for the synthesis of these compounds are described, and structure-activity relationships among the series are analyzed. In addition, initial findings regarding the mechanism of LF inhibition for these molecules are presented.
Collapse
Affiliation(s)
- Guan-Sheng Jiao
- Department of Chemistry, Hawaii Biotech, Inc., 99-193 Aiea Heights Dr., Suite 200, Aiea, 96701, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
The search for an MMP inhibitor with anticancer efficacy is a nearly three-decade endeavor. This inhibitor is yet to be found. The reasons for this failure include shortcomings in the chemistry of these compounds (including broad MMP sub-type selectivity, metabolic lability, and toxicity) as well as the emerging, and arguably extraordinary, complexity of MMP cell (and cancer) biology. Together these suggest that the successful anticancer inhibitor must possess MMP selectivity against the MMP subtype whose involvement is critical, yet highly temporally (with respect to metastatic progression) and mechanistically (with respect to matrix degradation) regulated. This review summarizes the progression of chemical structure and mechanistic thinking toward these objectives, with emphasis on the disappointment, the perseverance, and the resilient optimism that such an inhibitor is there to be discovered.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556-5670, USA
| | | |
Collapse
|
27
|
Jiao GS, Cregar L, Goldman ME, Millis SZ, Tang C. Guanidinylated 2,5-dideoxystreptamine derivatives as anthrax lethal factor inhibitors. Bioorg Med Chem Lett 2006; 16:1527-31. [PMID: 16386899 DOI: 10.1016/j.bmcl.2005.12.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2005] [Revised: 12/09/2005] [Accepted: 12/09/2005] [Indexed: 11/23/2022]
Abstract
Anthrax lethal factor is a Zn(2+)-dependent metalloprotease and the key virulence factor of tripartite anthrax toxin secreted by Bacillus anthracis, the causative agent of anthrax. A series of guanidinylated 2,5-dideoxystreptamine derivatives were designed and synthesized as inhibitors of lethal factor, some of which show strong inhibitory activity against lethal factor in an in vitro FRET assay. Preparation and structure-activity relationships of these compounds are presented.
Collapse
Affiliation(s)
- Guan-Sheng Jiao
- Department of Chemistry, Hawaii Biotech Inc., 99-193 Aiea Heights Drive, Suite 200, Aiea, HI 96701, USA.
| | | | | | | | | |
Collapse
|