1
|
O’Shaughnessy M, Sheils O, Baird AM. The Lung Microbiome in COPD and Lung Cancer: Exploring the Potential of Metal-Based Drugs. Int J Mol Sci 2023; 24:12296. [PMID: 37569672 PMCID: PMC10419288 DOI: 10.3390/ijms241512296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer 17 are two of the most prevalent and debilitating respiratory diseases worldwide, both associated with high morbidity and mortality rates. As major global health concerns, they impose a substantial burden on patients, healthcare systems, and society at large. Despite their distinct aetiologies, lung cancer and COPD share common risk factors, clinical features, and pathological pathways, which have spurred increasing research interest in their co-occurrence. One area of particular interest is the role of the lung microbiome in the development and progression of these diseases, including the transition from COPD to lung cancer. Exploring novel therapeutic strategies, such as metal-based drugs, offers a potential avenue for targeting the microbiome in these diseases to improve patient outcomes. This review aims to provide an overview of the current understanding of the lung microbiome, with a particular emphasis on COPD and lung cancer, and to discuss the potential of metal-based drugs as a therapeutic strategy for these conditions, specifically concerning targeting the microbiome.
Collapse
Affiliation(s)
- Megan O’Shaughnessy
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, St. James’s Hospital, D08 RX0X Dublin, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
| |
Collapse
|
2
|
O’Shaughnessy M, Piatek M, McCarron P, McCann M, Devereux M, Kavanagh K, Howe O. In Vivo Activity of Metal Complexes Containing 1,10-Phenanthroline and 3,6,9-Trioxaundecanedioate Ligands against Pseudomonas aeruginosa Infection in Galleria mellonella Larvae. Biomedicines 2022; 10:biomedicines10020222. [PMID: 35203432 PMCID: PMC8869450 DOI: 10.3390/biomedicines10020222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022] Open
Abstract
Drug-resistant Pseudomonas aeruginosa is rapidly developing resulting in a serious global threat. Immunocompromised patients are specifically at risk, especially those with cystic fibrosis (CF). Novel metal complexes incorporating 1,10-phenanthroline (phen) ligands have previously demonstrated antibacterial and anti-biofilm effects against resistant P. aeruginosa from CF patients in vitro. Herein, we present the in vivo efficacy of {[Cu(3,6,9-tdda)(phen)2]·3H2O·EtOH}n (Cu-tdda-phen), {[Mn(3,6,9-tdda)(phen)2]·3H2O·EtOH}n (Mn-tdda-phen) and [Ag2(3,6,9-tdda)(phen)4]·EtOH (Ag-tdda-phen) (tddaH2 = 3,6,9-trioxaundecanedioic acid). Individual treatments of these metal-tdda-phen complexes and in combination with the established antibiotic gentamicin were evaluated in vivo in larvae of Galleria mellonella infected with clinical isolates and laboratory strains of P. aeruginosa. G. mellonella were able to tolerate all test complexes up to 10 µg/larva. In addition, the immune response was affected by stimulation of immune cells (hemocytes) and genes that encode for immune-related peptides, specifically transferrin and inducible metallo-proteinase inhibitor. The amalgamation of metal-tdda-phen complexes and gentamicin further intensified this response at lower concentrations, clearing a P. aeruginosa infection that were previously resistant to gentamicin alone. Therefore this work highlights the anti-pseudomonal capabilities of metal-tdda-phen complexes alone and combined with gentamicin in an in vivo model.
Collapse
Affiliation(s)
- Megan O’Shaughnessy
- School of Biological and Health Sciences, Technological University Dublin-City Campus, D07 ADY7 Dublin, Ireland;
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
| | - Magdalena Piatek
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, W23 F2H6 Kildare, Ireland;
| | - Pauraic McCarron
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
| | - Malachy McCann
- Chemistry Department, Maynooth University, W23 F2H6 Kildare, Ireland;
| | - Michael Devereux
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
| | - Kevin Kavanagh
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, W23 F2H6 Kildare, Ireland;
- Correspondence: (K.K.); (O.H.)
| | - Orla Howe
- School of Biological and Health Sciences, Technological University Dublin-City Campus, D07 ADY7 Dublin, Ireland;
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
- Correspondence: (K.K.); (O.H.)
| |
Collapse
|
3
|
Kasyanenko N, Qiushi Z, Bakulev V, Osolodkov M, Sokolov P, Demidov V. DNA Binding with Acetate Bis(1,10-phenanthroline)silver(I) Monohydrate in a Solution and Metallization of Formed Structures. Polymers (Basel) 2017; 9:E211. [PMID: 30970890 PMCID: PMC6432125 DOI: 10.3390/polym9060211] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 02/07/2023] Open
Abstract
The study of DNA interaction with the acetate bis(1,10-phenanthroline)silver(I) monohydrate in a solution is of interest both for understanding the mechanism of biological activity of silver compound and for forming ordered structures (DNA fibrils) that can be used to solve various problems in the field of nanotechnology. The analysis of changing the DNA conformation (secondary structure, persistent length and volume effects) during the interaction by the methods of UV spectroscopy with the analysis of DNA melting, circular dichroism, viscosity, flow birefringence, AFM (atomic force microscopy) and SEM (scanning electron microscopy) was performed. The formation of two types of complexes was observed. At lower concentration of compound in DNA solution, silver atoms form the coordination bonds with a macromolecule, while the released phenanthroline ligands intercalate between DNA bases. When the concentration of the compound increases, the phenanthroline ligands form an ordered "layer" around the helix. The excess of silver compounds in the DNA solution (with more than five silver atoms per base pair), DNA precipitation is observed with the formation of long fibrils. It was shown that the binding of silver to DNA during the formation of complexes provides further metallization of the resulting structures with the aid of reducing agents; phenanthroline ligands influence the result of such metallization.
Collapse
Affiliation(s)
- Nina Kasyanenko
- Department of Physics, St. Petersburg State University, Universitetskaya Naberezhnaya 3/7, 199037 St. Petersburg, Russia.
| | - Zhang Qiushi
- Department of Physics, St. Petersburg State University, Universitetskaya Naberezhnaya 3/7, 199037 St. Petersburg, Russia.
| | - Vladimir Bakulev
- Department of Physics, St. Petersburg State University, Universitetskaya Naberezhnaya 3/7, 199037 St. Petersburg, Russia.
| | - Mikhail Osolodkov
- Department of Physics, St. Petersburg State University, Universitetskaya Naberezhnaya 3/7, 199037 St. Petersburg, Russia.
| | - Petr Sokolov
- Department of Physics, St. Petersburg State University, Universitetskaya Naberezhnaya 3/7, 199037 St. Petersburg, Russia.
| | - Viktor Demidov
- Pro-Brite Company, Sofiyskaya ul., 93, 192289 St. Petersburg, Russia.
| |
Collapse
|
4
|
Pommier Y, Pilon A, Bajaj K, Mazumder A, Neamati N. HIV-1 Integrase as a Target for Antiviral Drugs. ACTA ACUST UNITED AC 2017. [DOI: 10.1177/095632029700800601] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Y Pommier
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, Building 37, Room 5C25, National Institutes of Health, Bethesda, MD 20892-4255, USA
| | - Aa Pilon
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, Building 37, Room 5C25, National Institutes of Health, Bethesda, MD 20892-4255, USA
| | - K Bajaj
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, Building 37, Room 5C25, National Institutes of Health, Bethesda, MD 20892-4255, USA
| | - A Mazumder
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, Building 37, Room 5C25, National Institutes of Health, Bethesda, MD 20892-4255, USA
| | - N Neamati
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, Building 37, Room 5C25, National Institutes of Health, Bethesda, MD 20892-4255, USA
| |
Collapse
|
5
|
Chitra L, Kumar CR, Basha HM, Ponne S, Boopathy R. Interaction of metal chelators with different molecular forms of acetylcholinesterase and its significance in Alzheimer's disease treatment. Proteins 2013; 81:1179-91. [DOI: 10.1002/prot.24267] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 01/22/2013] [Accepted: 02/06/2013] [Indexed: 01/31/2023]
Affiliation(s)
- Loganathan Chitra
- DRDO-BU Center for Life Sciences; Bharathiar University; Coimbatore; 641 046; Tamil Nadu; India
| | - Chinnadurai Raj Kumar
- Department of Biotechnology; School of Biotechnology and Genetic Engineering; Bharathiar University; Coimbatore; 641 046; Tamil Nadu; India
| | - Haleema M. Basha
- Department of Biotechnology; School of Biotechnology and Genetic Engineering; Bharathiar University; Coimbatore; 641 046; Tamil Nadu; India
| | - Saravanaraman Ponne
- Department of Biotechnology; School of Biotechnology and Genetic Engineering; Bharathiar University; Coimbatore; 641 046; Tamil Nadu; India
| | | |
Collapse
|
6
|
Demidov VN, Kas’yanenko NA, Antonov VS, Volkov IL, Sokolov PA, Pakhomova TB, Simanova SA. Reaction with DNA and pharmacologic activity of 1,10-phenanthroline and electron-rich 1,10-phenanthrocyanine complexes of d-elements. RUSS J GEN CHEM+ 2012. [DOI: 10.1134/s1070363212030401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
7
|
HIV‐1 Integrase Inhibitors: Update and Perspectives. HIV-1: MOLECULAR BIOLOGY AND PATHOGENESIS 2008; 56:199-228. [DOI: 10.1016/s1054-3589(07)56007-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
8
|
Nefzi A, Hoesl CE, Pinilla C, Kauffman GB, Maggiora GM, Pasquale E, Houghten RA. Synthesis of Platinum(II) Chiral Tetraamine Coordination Complexes. ACTA ACUST UNITED AC 2006; 8:780-3. [PMID: 16961414 DOI: 10.1021/cc060045h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The individual and combinatorial syntheses of individual as well as a mixture-based diversity of 195 112 platinum(II) coordination complexes of chiral tetraamines are described. The use of both solid-phase synthesis and solution phase follow-on approaches were found to best afford the title compounds.
Collapse
Affiliation(s)
- Adel Nefzi
- Torrey Pines Institute for Molecular Studies, 3550 General Atomics Court, San Diego, California 92121, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Smolov M, Gottikh M, Tashlitskii V, Korolev S, Demidyuk I, Brochon JC, Mouscadet JF, Deprez E. Kinetic study of the HIV-1 DNA 3'-end processing. FEBS J 2006; 273:1137-51. [PMID: 16519680 DOI: 10.1111/j.1742-4658.2006.05139.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The 3'-processing of viral DNA extremities is the first step in the integration process catalysed by human immunodeficiency virus (HIV)-1 integrase (IN). This reaction is relatively inefficient and processed DNAs are usually detected in vitro under conditions of excess enzyme. Despite such experimental conditions, steady-state Michaelis-Menten formalism is often applied to calculate characteristic equilibrium/kinetic constants of IN. We found that the amount of processed product was not significantly affected under conditions of excess DNA substrate, indicating that IN has a limited turnover for DNA cleavage. Therefore, IN works principally in a single-turnover mode and is intrinsically very slow (single-turnover rate constant = 0.004 min(-1)), suggesting that IN activity is mainly limited at the chemistry step or at a stage that precedes chemistry. Moreover, fluorescence experiments showed that IN-DNA product complexes were very stable over the time-course of the reaction. Binding isotherms of IN to DNA substrate and product also indicate tight binding of IN to the reaction product. Therefore, the slow cleavage rate and limited product release prevent or greatly reduce subsequent turnover. Nevertheless, the time-course of product formation approximates to a straight line for 90 min (apparent initial velocity), but we show that this linear phase is due to the slow single-turnover rate constant and does not indicate steady-state multiple turnover. Finally, our data ruled out the possibility that there were large amounts of inactive proteins or dead-end complexes in the assay. Most of complexes initially formed were active although dramatically slow.
Collapse
Affiliation(s)
- Maksim Smolov
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Russia
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Human immunodeficiency virus Type 1 (HIV-1) integrase is an essential enzyme for the obligatory integration of the viral DNA into the infected cell chromosome. As no cellular homologue of HIV integrase has been identified, this unique HIV-1 enzyme is an attractive target for the development of new therapeutics. Treatment of HIV-1 infection and AIDS currently consists of the use of combinations of HIV-1 inhibitors directed against reverse transcriptase (RT) and protease. However, their numerous side effects and the rapid emergence of drug-resistant variants limit greatly their use in many AIDS patients. In principle, inhibitors of the HIV-1 integrase should be relatively non-toxic and provide additional benefits for AIDS chemotherapy. There have been many major advances in our understanding of the molecular mechanism of the integration reaction, although some critical aspects remain obscure. Several classes of compounds have been screened and further scrutinised for their inhibitory properties against the HIV integrase; however, there are currently no useful inhibitors available clinically for the treatment of AIDS patients. This review describes the current knowledge of the biological functions of the HIV-1 integrase and reports the major classes of integrase inhibitors identified to date.
Collapse
Affiliation(s)
- Khampoune Sayasith
- CRRA, Faculty of Veterinary Medicine, University of Montreal, PO Box 5000, St-Hyacinthe, Quebec, Canada J2S 7C6.
| | | | | |
Collapse
|
11
|
Neamati N, Marchand C, Pommier Y. HIV-1 integrase inhibitors: past, present, and future. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2001; 49:147-65. [PMID: 11013763 DOI: 10.1016/s1054-3589(00)49026-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Affiliation(s)
- N Neamati
- Laboratory of Molecular Pharmacology, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
12
|
Molteni V, Rhodes D, Rubins K, Hansen M, Bushman FD, Siegel JS. A new class of HIV-1 integrase inhibitors: the 3,3,3', 3'-tetramethyl-1,1'-spirobi(indan)-5,5',6,6'-tetrol family. J Med Chem 2000; 43:2031-9. [PMID: 10821715 DOI: 10.1021/jm990600c] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Integration is a required step in HIV replication, but as yet no inhibitors of the integration step have been developed for clinical use. Many inhibitors have been identified that are active against purified viral-encoded integrase protein; of these, many contain a catechol moiety. Though this substructure contributes potency in inhibitors, it is associated with toxicity and so the utility of catechol-containing inhibitors has been questioned. We have synthesized and tested a systematic series of derivatives of a catechol-containing inhibitor (1) with the goal of identifying catechol isosteres that support inhibition. We find that different patterns of substitution on the aromatic ring suffice for inhibition when Mn(2+) is used as a cofactor. Importantly, the efficiency is different when Mg(2+), the more likely in vivo cofactor, is used. These data emphasize the importance of assays with Mg(2+) and offer new catechol isosteres for use in integrase inhibitors.
Collapse
Affiliation(s)
- V Molteni
- Department of Chemistry, University of California, San Diego, La Jolla, California 92093-0358, USA
| | | | | | | | | | | |
Collapse
|
13
|
Affiliation(s)
- A Y Louie
- Department of Biology, Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, California 91125
| | | |
Collapse
|
14
|
Abstract
Integration of the viral DNA into a host cell chromosome is an essential step for HIV replication and maintenance of persistent infection. Two viral factors are essential for integration: the viral DNA termini (the att sites) and IN. Accruing knowledge of the IN structure, catalytic mechanisms, and interactions with other proteins can be used to design strategies to block integration. A large number of inhibitors have been identified that can be used as leads for the development of potent and selective anti-IN drugs with antiviral activity.
Collapse
Affiliation(s)
- Y Pommier
- Laboratory of Molecular Pharmacology, National Cancer Institute, Bethesda, Maryland 20892-4255, USA
| | | |
Collapse
|
15
|
Reddy MV, Rao MR, Rhodes D, Hansen MS, Rubins K, Bushman FD, Venkateswarlu Y, Faulkner DJ. Lamellarin alpha 20-sulfate, an inhibitor of HIV-1 integrase active against HIV-1 virus in cell culture. J Med Chem 1999; 42:1901-7. [PMID: 10354398 DOI: 10.1021/jm9806650] [Citation(s) in RCA: 229] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
HIV-1 integrase is an attractive target for anti-retroviral chemotherapy, but to date no clinically useful inhibitors have been developed. We have screened diverse marine natural products for compounds active against integrase in vitro and found a series of ascidian alkaloids, the lamellarins, that show selective inhibition. A new member of the family named lamellarin alpha 20-sulfate (1), the structure of which was determined from spectroscopic data, displayed the most favorable therapeutic index. The site of action of lamellarin alpha 20-sulfate on the integrase protein was mapped by testing activity against deletion mutants of integrase. Inhibition of isolated catalytic domain was detectable though weaker than inhibition of full length integrase; possibly lamellarin alpha 20-sulfate binds a site composed of multiple integrase domains. Lamellarin alpha 20-sulfate also inhibited integration in vitro by authentic HIV-1 replication intermediates isolated from infected cells. Lamellarin alpha 20-sulfate was tested against wild type HIV using the MAGI indicator cell assay and found to inhibit early steps of HIV replication. To clarify the inhibitor target, we tested inhibition against an HIV-based retroviral vector bearing a different viral envelope. Inhibition was observed, indicating that the HIV envelope cannot be the sole target of lamellarin alpha 20-sulfate in cell culture. In addition, these single round tests rule out action against viral assembly or budding. These findings provide a new class of compounds for potential development of clinically useful integrase inhibitors.
Collapse
Affiliation(s)
- M V Reddy
- Organic Chemistry Division-I, Natural Products Laboratory, Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Hazuda D, Blau CU, Felock P, Hastings J, Pramanik B, Wolfe A, Bushman F, Farnet C, Goetz M, Williams M, Silverman K, Lingham R, Singh S. Isolation and characterization of novel human immunodeficiency virus integrase inhibitors from fungal metabolites. Antivir Chem Chemother 1999; 10:63-70. [PMID: 10335400 DOI: 10.1177/095632029901000202] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
We have identified a series of novel inhibitors of human immunodeficiency virus type 1 (HIV-1) integrase by randomly screening natural product extracts using an in vitro biochemical assay designed to identify inhibitors of integrase-catalysed strand transfer. Equisetin recovered from the fungus Fusarium heterosporum and a novel enantiomeric homologue of equisetin from Phoma sp. were isolated as inhibitors of HIV-1 integrase in vitro. Two additional analogues, a novel decalin derivative, integric acid, and oteromycin were also discovered to be inhibitors of integrase. Equisetin and related compounds inhibit 3' end-processing and strand transfer as well as disintegration catalysed by either the full-length enzyme or the truncated integrase core domain (amino acids 50-212). These compounds also inhibit strand transfer reactions catalysed by stable complexes assembled in vitro and integration reactions catalysed by pre-integration complexes isolated from HIV-1-infected cells. The compounds described in this report are structurally novel and mechanistically distinct from many previously described inhibitors of HIV-1 integrase. These results demonstrate the utility of using an appropriately configured assay to identify compounds that are effective post-assembly and the potential of isolating novel integrase inhibitors from complex natural product extracts.
Collapse
Affiliation(s)
- D Hazuda
- Department of Antiviral Research, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Farnet CM, Wang B, Hansen M, Lipford JR, Zalkow L, Robinson WE, Siegel J, Bushman F. Human immunodeficiency virus type 1 cDNA integration: new aromatic hydroxylated inhibitors and studies of the inhibition mechanism. Antimicrob Agents Chemother 1998; 42:2245-53. [PMID: 9736543 PMCID: PMC105796 DOI: 10.1128/aac.42.9.2245] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Integration of the human immunodeficiency virus type 1 (HIV-1) cDNA is a required step for viral replication. Integrase, the virus-encoded enzyme important for integration, has not yet been exploited as a target for clinically useful inhibitors. Here we report on the identification of new polyhydroxylated aromatic inhibitors of integrase including ellagic acid, purpurogallin, 4,8, 12-trioxatricornan, and hypericin, the last of which is known to inhibit viral replication. These compounds and others were characterized in assays with subviral preintegration complexes (PICs) isolated from HIV-1-infected cells. Hypericin was found to inhibit PIC assays, while the other compounds tested were inactive. Counterscreening of these and other integrase inhibitors against additional DNA-modifying enzymes revealed that none of the polyhydroxylated aromatic compounds are active against enzymes that do not require metals (methylases, a pox virus topoisomerase). However, all were cross-reactive with metal-requiring enzymes (restriction enzymes, a reverse transcriptase), implicating metal atoms in the inhibitory mechanism. In mechanistic studies, we localized binding of some inhibitors to the catalytic domain of integrase by assaying competition of binding by labeled nucleotides. These findings help elucidate the mechanism of action of the polyhydroxylated aromatic inhibitors and provide practical guidance for further inhibitor development.
Collapse
Affiliation(s)
- C M Farnet
- Salk Institute for Biological Studies, La Jolla, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Hansen MS, Carteau S, Hoffmann C, Li L, Bushman F. Retroviral cDNA integration: mechanism, applications and inhibition. GENETIC ENGINEERING 1998; 20:41-61. [PMID: 9666555 DOI: 10.1007/978-1-4899-1739-3_3] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- M S Hansen
- Infectious Disease Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
19
|
Gerton JL, Ohgi S, Olsen M, DeRisi J, Brown PO. Effects of mutations in residues near the active site of human immunodeficiency virus type 1 integrase on specific enzyme-substrate interactions. J Virol 1998; 72:5046-55. [PMID: 9573274 PMCID: PMC110067 DOI: 10.1128/jvi.72.6.5046-5055.1998] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The phylogenetically conserved catalytic core domain of human immunodeficiency virus type 1 (HIV-1) integrase contains elements necessary for specific recognition of viral and target DNA features. In order to identify specific amino acids that determine substrate specificity, we mutagenized phylogenetically conserved residues that were located in close proximity to the active-site residues in the crystal structure of the isolated catalytic core domain of HIV-1 integrase. Residues composing the phylogenetically conserved DD(35)E active-site motif were also mutagenized. Purified mutant proteins were evaluated for their ability to recognize the phylogenetically conserved CA/TG base pairs near the viral DNA ends and the unpaired dinucleotide at the 5' end of the viral DNA, using disintegration substrates. Our findings suggest that specificity for the conserved A/T base pair depends on the active-site residue E152. The phenotype of IN(Q148L) suggested that Q148 may be involved in interactions with the 5' dinucleotide of the viral DNA end. The activities of some of the proteins with mutations in residues in close proximity to the active-site aspartic and glutamic acids were salt sensitive, suggesting that these mutations disrupted interactions with DNA.
Collapse
Affiliation(s)
- J L Gerton
- Department of Microbiology and Immunology, Stanford University Medical Center, Stanford, California 94305-5428, USA
| | | | | | | | | |
Collapse
|
20
|
Dolgounitcheva O, Zakrzewski VG, Ortiz JV. Ionization Energies of Acridine, Phenazine, and Diazaphenanthrenes. J Phys Chem A 1997. [DOI: 10.1021/jp971910e] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- O. Dolgounitcheva
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506-3701
| | - V. G. Zakrzewski
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506-3701
| | - J. V. Ortiz
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506-3701
| |
Collapse
|
21
|
|
22
|
Hazuda DJ, Felock PJ, Hastings JC, Pramanik B, Wolfe AL. Differential divalent cation requirements uncouple the assembly and catalytic reactions of human immunodeficiency virus type 1 integrase. J Virol 1997; 71:7005-11. [PMID: 9261430 PMCID: PMC191986 DOI: 10.1128/jvi.71.9.7005-7011.1997] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Previous in vitro analyses have shown that the human immunodeficiency virus type 1 (HIV-1) integrase uses either manganese or magnesium to assemble as a stable complex on the donor substrate and to catalyze strand transfer. We now demonstrate that subsequent to assembly, catalysis of both 3' end processing and strand transfer requires a divalent cation cofactor and that the divalent cation requirements for assembly and catalysis can be functionally distinguished based on the ability to utilize calcium and cobalt, respectively. The different divalent cation requirements manifest by these processes are exploited to uncouple assembly and catalysis, thus staging the reaction. Staged 3' end processing and strand transfer assays are then used in conjunction with exonuclease III protection analysis to investigate the effects of integrase inhibitors on each step in the reaction. Analysis of a series of related inhibitors demonstrates that these types of compounds affect assembly and not either catalytic process, therefore reconciling the apparent disparate results obtained for such inhibitors in assays using isolated preintegration complexes. These studies provide evidence for a distinct role of the divalent cation cofactor in assembly and catalysis and have implications for both the identification and characterization of integrase inhibitors.
Collapse
Affiliation(s)
- D J Hazuda
- Department of Antiviral Research, Merck Research Laboratories, West Point, Pennsylvania 19486, USA
| | | | | | | | | |
Collapse
|
23
|
Mazumder A, Uchida H, Neamati N, Sunder S, Jaworska-Maslanka M, Wickstrom E, Zeng F, Jones RA, Mandes RF, Chenault HK, Pommier Y. Probing interactions between viral DNA and human immunodeficiency virus type 1 integrase using dinucleotides. Mol Pharmacol 1997; 51:567-75. [PMID: 9106620 DOI: 10.1124/mol.51.4.567] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Retroviral integrases are essential for viral replication and represent an attractive chemotherapeutic target. In the current study, we demonstrated the activity of micromolar concentrations of dinucleotides against human immunodeficiency virus type 1 (HIV-1), HIV type 2 (HIV-2), simian immunodeficiency virus, and feline immunodeficiency virus integrases. The structure-activity relationship indicates that 5'-phosphorylation enhances potency and that phosphodiester and sugar modifications affect the inhibition of HIV-1 integrase. Base sequence selectivity was observed: pAC, pAT, and pCT were the most potent inhibitors, whereas pAA, pGA, and pGC showed low activity at 100 microM. The inhibition by pAC is consistent with the interaction of the enzyme with the 5' end of the noncleaved strand (5'-AC-3'). The linear and cyclic dinucleotides released by the 3'-processing reaction did not affect enzymatic activity at physiological concentrations. An increase in the length to trinucleotides or tetranucleotides enhanced potency by only 2-3-fold, suggesting that two neighboring bases may be sufficient for significant interactions. Inhibition of a truncated (50-212) integrase mutant and global inhibition of all nucleophiles in the 3'-processing reaction suggest that dinucleotides bind in the catalytic core. All of the active dinucleotides inhibited enzyme/DNA binding in their respective IC50 range. Although the dinucleotides tested showed no antiviral activity, these observations demonstrate the usefulness of dinucleotides in elucidating enzyme mechanisms and as potential ligands for cocrystallization and as lead structures for development of antivirals.
Collapse
Affiliation(s)
- A Mazumder
- Division of Basic Sciences, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Hong H, Neamati N, Wang S, Nicklaus MC, Mazumder A, Zhao H, Burke TR, Pommier Y, Milne GW. Discovery of HIV-1 integrase inhibitors by pharmacophore searching. J Med Chem 1997; 40:930-6. [PMID: 9083481 DOI: 10.1021/jm960754h] [Citation(s) in RCA: 81] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Based upon a class of known HIV-1 integrase inhibitors, several pharmacophore models were proposed from molecular modeling studies and validated using a 3D database of 152, compounds for which integrase assay data are known. Using the most probable pharmacophore model as the query, the NCI 3D database of 206,876 compounds was searched, and 340 compounds that contain the pharmacophore query were identified. Twenty-nine of these compounds were selected and tested in the HIV-1 integrase assay. This led to the discovery of 10 novel, structurally diverse HIV-1 integrase inhibitors, four of which have an IC50 value less than 30 microM and are promising lead compounds for further HIV-1 integrase inhibitor development.
Collapse
Affiliation(s)
- H Hong
- Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4255, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Nicklaus MC, Neamati N, Hong H, Mazumder A, Sunder S, Chen J, Milne GW, Pommier Y. HIV-1 integrase pharmacophore: discovery of inhibitors through three-dimensional database searching. J Med Chem 1997; 40:920-9. [PMID: 9083480 DOI: 10.1021/jm960596u] [Citation(s) in RCA: 148] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Starting from a known inhibitor of human immunodeficiency virus type 1 (HIV-1) integrase (IN); caffeic acid phenethyl ester (CAPE), a putative three-point pharmacophore for binding of inhibitors to IN was derived. This pharmacophore was used to search the National Cancer Institute three-dimensional (3D) structural database. Out of the open, nonproprietary part of this database, comprising approximately 200000 compounds, 267 structures were found to match the pharmacophore in at least one conformation, and 60 of those were tested in an in vitro assay against HIV-1 IN. Out of these, 19 were found to inhibit both the 3'-processing and strand transfer of IN at micromolar concentrations. In order to test the validity of this pharmacophore, a small 3D database of 152 published IN inhibitors was built. A search in this database yielded a statistically significant correlation of the presence of this pharmacophore and the potency of the compounds. An automated pharmacophore identification procedure performed on this set of compounds provided additional support for the importance of this pharmacophore for binding of inhibitors to IN and hinted at a possible second pharmacophore. The role of aromatic moieties in the binding of ligands to HIV-1 IN through interactions with divalent metal cations, which are known to be necessary for activity of the enzyme, was explored in ab initio calculations.
Collapse
Affiliation(s)
- M C Nicklaus
- Laboratory of Medicinal Chemistry, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4255, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Hazuda D, Felock P, Hastings J, Pramanik B, Wolfe A, Goodarzi G, Vora A, Brackmann K, Grandgenett D. Equivalent inhibition of half-site and full-site retroviral strand transfer reactions by structurally diverse compounds. J Virol 1997; 71:807-11. [PMID: 8985421 PMCID: PMC191122 DOI: 10.1128/jvi.71.1.807-811.1997] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
In vitro assay systems which use recombinant retroviral integrase (IN) and short DNA oligonucleotides fail to recapitulate the full-site integration reaction as it is known to occur in vivo. The relevance of using such circumscribed in vitro assays to define inhibitors of retroviral integration has not been formerly demonstrated. Therefore, we analyzed a series of structurally diverse inhibitors with respect to inhibition of both half-site and full-site strand transfer reactions with either recombinant or virion-produced IN. Half-site and full-site reactions catalyzed by avian myeloblastosis virus and human immunodeficiency virus type 1 (HIV-1) IN from virions are shown to be equivalently sensitive to inhibition by compounds which inhibit half-site reactions catalyzed by the recombinant HIV-1 IN. These studies therefore support the utility of using in vitro assays employing either recombinant or virion-derived IN to identify inhibitors of integration.
Collapse
Affiliation(s)
- D Hazuda
- Department of Antiviral Research, Merck Research Laboratories, West Point, Pennsylvania 19486, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Farnet CM, Wang B, Lipford JR, Bushman FD. Differential inhibition of HIV-1 preintegration complexes and purified integrase protein by small molecules. Proc Natl Acad Sci U S A 1996; 93:9742-7. [PMID: 8790401 PMCID: PMC38499 DOI: 10.1073/pnas.93.18.9742] [Citation(s) in RCA: 103] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
To replicate, HIV-1 must integrate a cDNA copy of the viral RNA genome into a chromosome of the host. The integration system is a promising target for antiretroviral agents, but to date no clinically useful integration inhibitors have been identified. Previous screens for integrase inhibitors have assayed inhibition of reactions containing HIV-1 integrase purified from an Escherichia coli expression system. Here we compare action of inhibitors in vitro on purified integrase and on subviral preintegration complexes (PICs) isolated from lymphoid cells infected with HIV-1. We find that many inhibitors active against purified integrase are inactive against PICs. Using PIC assays as a primary screen, we have identified three new anthraquinone inhibitors active against PICs and also against purified integrase. We propose that PIC assays are the closest in vitro match to integration in vivo and, as such, are particularly appropriate for identifying promising integration inhibitors.
Collapse
Affiliation(s)
- C M Farnet
- Infectious Disease Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
28
|
Mazumder A, Wang S, Neamati N, Nicklaus M, Sunder S, Chen J, Milne GW, Rice WG, Burke TR, Pommier Y. Antiretroviral agents as inhibitors of both human immunodeficiency virus type 1 integrase and protease. J Med Chem 1996; 39:2472-81. [PMID: 8691444 DOI: 10.1021/jm960074e] [Citation(s) in RCA: 122] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The human immunodeficiency virus type one integrase (HIV-1 integrase) is required for integration of a double-stranded DNA copy of the viral RNA genome into a host chromosome and for HIV replication. We have previously reported that phenolic moieties in compounds such as flavones, caffeic acid phenethyl ester (CAPE), tyrphostins, and curcumin confer inhibitory activity against HIV-1 integrase. We have investigated the actions of several recently described protease inhibitors, possessing novel structural features, on HIV-1 integrase. NSC 158393, which contains four 4-hydroxycoumarin residues, was found to exhibit antiviral, antiprotease, and antiintegrase activity. Both the DNA binding and catalytic activities (3'-processing and strand transfer) of integrase were inhibited at micromolar concentrations. Disintegration catalyzed by an integrase mutant containing only the central catalytic domain was also inhibited, indicating that the binding site for these compounds resides in the central 50-212 amino acids of HIV-1 integrase. Binding at or near the integrase catalytic site was also suggested by a global inhibition of the choice of attacking nucleophile in the 3'-processing reaction. NSC 158393 inhibited HIV-2, feline, and simian immunodeficiency virus integrases while eukaryotic topoisomerase I was inhibited at higher concentrations, suggesting selective inhibition of retroviral integrases. Molecular modeling studies revealed that the two hydroxyls and two carbonyl moieties in NSC 158393 may represent essential elements of the pharmacophore. Antiviral efficacy was observed with NSC 158393 derivatives that inhibited both HIV protease and integrase, and the most potent integrase inhibitors also inhibited HIV protease. Hydroxycoumarins may provide lead compounds for development of novel antiviral agents based upon the concurrent inhibition of two viral targets, HIV-1 integrase and protease.
Collapse
Affiliation(s)
- A Mazumder
- Laboratory of Molecular Pharmacology, Division of Basic Sciences, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Bouziane M, Cherny DI, Mouscadet JF, Auclair C. Alternate strand DNA triple helix-mediated inhibition of HIV-1 U5 long terminal repeat integration in vitro. J Biol Chem 1996; 271:10359-64. [PMID: 8626607 DOI: 10.1074/jbc.271.17.10359] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Integration of the human immunodeficiency virus (HIV) DNA into the host genome is an obligatory process in the replicative life cycle of the virus. This event is mediated in vitro by integrase, a viral protein which binds to specific sequences located on both extremities of the DNA long terminal repeats (LTRs). These sites are highly conserved in all HIV genomes and thus provide potential targets for the selective inhibition of integration. The integrase-binding site located on the HIV-1 U5 LTR end contains two adjacent purine tracts on opposite strands, 5' . . . GGAAAATCTCT-3'/3'-CCTTTTAGAGA . . . 5', in parallel orientations. A single strand oligonucleotide 5'-GGTTTTTGTGT-3' was designed to associate with these tracts via its ability to form a continuous alternate strand DNA triplex. Under neutral pH and physiological temperature, the oligonucleotide, tagged with an intercalator chromophore oxazolopyridocarbazole, formed a stable triplex with the target DNA. The occurrence of this unusual triplex was demonstrated by both DNase I footprinting and electron microscopy. The triplex inhibits the two steps of the integrase-mediated reactions, namely, the endonucleolytic cleavage of the dinucleotide 5'-GT-3' from the 3' end of the integration substrate and the integration of the substrate into the heterologous target DNA. The midpoints for both inhibition reactions were observed at oligonucleotide concentrations of 50-100 nM. We believe that these results open new possibilities for the specific targeting of viral DNA LTR ends with the view of inhibiting integration under physiological conditions.
Collapse
Affiliation(s)
- M Bouziane
- Laboratoire de Physicochimie et Pharmacologie des Macromolécules Biologiques CNRS URA 147, Institut Gustave Roussy, Rue Camille Desmoulins, 94805 Villejuif, France
| | | | | | | |
Collapse
|
30
|
Eich E, Pertz H, Kaloga M, Schulz J, Fesen MR, Mazumder A, Pommier Y. (-)-Arctigenin as a lead structure for inhibitors of human immunodeficiency virus type-1 integrase. J Med Chem 1996; 39:86-95. [PMID: 8568830 DOI: 10.1021/jm950387u] [Citation(s) in RCA: 151] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The natural dibenzylbutyrolactone type lignanolide (-)-arctigenin (2), an inhibitor of human immunodeficiency virus type-1 (HIV-1) replication in infected human cell systems, was found to suppress the integration of proviral DNA into the cellular DNA genome. In the present study 2 was tested with purified HIV-1 integrase and found to be inactive in the cleavage (3'-processing) and integration (strand transfer) assays. However, the semisynthetic 3-O-demethylated congener 9 characterized by a catechol substructure exhibited remarkable activities in both assays. Structure-activity relationship studies with 30 natural (1-6), semisynthetic (7-21), and synthetic (37-43, 45, 46) lignans revealed that (1) the lactone moiety is crucial since compounds with a butane-1,4-diol or tetrahydrofuran substructure and also lignanamide analogues lacked activity and (2) the number and arrangement of phenolic hydroxyl groups is important for the activity of lignanolides. The congener with two catechol substructures (7) was found to be the most active compound in this study. 7 was also a potent inhibitor of the "disintegration" reaction which models the reversal of the strand transfer reaction. The inhibitory activity of 7 with the core enzyme fragment consisting of amino acids 50-212 suggests that the binding site of 7 resides in the catalytic domain.
Collapse
Affiliation(s)
- E Eich
- Institut für Pharmazeutische Biologie, Freie Universität Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
31
|
Puras Lutzke RA, Eppens NA, Weber PA, Houghten RA, Plasterk RH. Identification of a hexapeptide inhibitor of the human immunodeficiency virus integrase protein by using a combinatorial chemical library. Proc Natl Acad Sci U S A 1995; 92:11456-60. [PMID: 8524782 PMCID: PMC40420 DOI: 10.1073/pnas.92.25.11456] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Integration of human immunodeficiency virus (HIV) DNA into the human genome requires the virus-encoded integrase (IN) protein, and therefore the IN protein is a suitable target for antiviral strategies. To find a potent HIV IN inhibitor, we screened a "synthetic peptide combinatorial library." We identified a hexapeptide with the sequence HCKFWW that inhibits IN-mediated 3'-processing and integration with an IC50 of 2 microM. The peptide is active on IN proteins from other retroviruses such as HIV-2, feline immunodeficiency virus, and Moloney murine leukemia virus, supporting the notion that a conserved region of IN is targeted. The hexapeptide was also tested in the disintegration reaction. This phosphoryl-transfer reaction can be carried out by the catalytic core of IN alone, and the peptide HCKFWW was found to inhibit this reaction, suggesting that the hexapeptide acts at or near the catalytic site of IN. Identification of an IN hexapeptide inhibitor provides proof of concept for the approach, and, moreover, this peptide may be useful for structure-function analysis of IN.
Collapse
Affiliation(s)
- R A Puras Lutzke
- Division of Molecular Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|