1
|
Wu CC, Chen MS, Lee TY, Cheng YJ, Tsou HH, Huang TS, Cho DY, Chen JY. Screening and identification of emodin as an EBV DNase inhibitor to prevent its biological functions. Virol J 2023; 20:148. [PMID: 37443068 PMCID: PMC10339607 DOI: 10.1186/s12985-023-02107-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND The Epstein-Barr virus (EBV) is a prevalent oncovirus associated with a variety of human illnesses. BGLF5, an EBV DNase with alkaline nuclease (AN) activity, plays important roles in the viral life cycle and progression of human malignancies and has been suggested as a possible diagnostic marker and target for cancer therapy. Methods used conventionally for the detection of AN activity, radioactivity-based nuclease activity assay and DNA digestion detection by gel electrophoresis, are not suitable for screening AN inhibitors; the former approach is unsafe, and the latter is complicated. In the present study, a fluorescence-based nuclease activity assay was used to screen several natural compounds and identify an EBV DNase inhibitor. RESULTS Fluorescence-based nuclease activity assays, in which the DNA substrate is labelled with PicoGreen dye, are cheaper, safer, and easier to perform. Herein, the results of the fluorescence-based nuclease activity assay were consistent with the results of the two conventional methods. In addition, the PicoGreen-labelling method was applied for the biochemical characterisation of viral nucleases. Using this approach, we explored EBV DNase inhibitors. After several rounds of screening, emodin, an anthraquinone derivative, was found to possess significant anti-EBV DNase activity. We verified the efficacy of emodin using the conventional DNA-cleavage assay. Furthermore, using comet assay and micronucleus formation detection, we confirmed that emodin can inhibit DNase-induced DNA damage and genomic instability. Additionally, emodin treatment inhibited EBV production. CONCLUSIONS Using a PicoGreen-mediated nuclease activity assay, we successfully demonstrated that emodin has the potential to inhibit EBV DNase nuclease activity. Emodin also inhibits EBV DNase-related biological functions, suggesting that it is a potential inhibitor of EBV DNase.
Collapse
Affiliation(s)
- Chung-Chun Wu
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, No. 2, Yude Rd., North Dist, Taichung City, 40447, Taiwan.
| | - Mei-Shu Chen
- National Institute of Cancer Research, National Health Research Institutes, No.35, Keyan Road, Zhunan Town, Miaoli County, Taiwan
| | - Ting-Ying Lee
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, No. 2, Yude Rd., North Dist, Taichung City, 40447, Taiwan
| | - Yu-Jhen Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Hsiao-Hui Tsou
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Tze-Sing Huang
- National Institute of Cancer Research, National Health Research Institutes, No.35, Keyan Road, Zhunan Town, Miaoli County, Taiwan
| | - Der-Yang Cho
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, No. 2, Yude Rd., North Dist, Taichung City, 40447, Taiwan
| | - Jen-Yang Chen
- National Institute of Cancer Research, National Health Research Institutes, No.35, Keyan Road, Zhunan Town, Miaoli County, Taiwan.
| |
Collapse
|
2
|
Lum KY, Taki AC, Gasser RB, Tietjen I, Ekins MG, White JM, Addison RS, Hayes S, St John J, Davis RA. Comatulins A-E, Taurine-Conjugated Anthraquinones from the Australian Crinoid Comatula rotalaria. JOURNAL OF NATURAL PRODUCTS 2020; 83:1971-1979. [PMID: 32478519 DOI: 10.1021/acs.jnatprod.0c00267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Chemical investigations of two specimens of the Australian crinoid Comatula rotalaria afforded five new taurine-conjugated anthraquinones, comatulins A-E (1-5), together with 11 known marine natural products (6-16). The chemical structures of all the compounds were elucidated by detailed spectroscopic and spectrometric data analysis. The first X-ray crystal structure of a crinoid-derived acyl anthraquinone, rhodocomatulin 5,7-dimethyl ether (8), is reported here. Compounds 1, 2, 6-13, and two additional naphthopyrone derivatives, 17 and 18, were evaluated for their ability to inhibit HIV-1 replication in vitro; none of the compounds were active at 100 μM. Furthermore, compounds 1, 2, 6-10, 14, 15, 17, and 18 were screened for nematocidal activity against exsheathed third-stage larvae of Hemonchus contortus, a highly pathogenic parasite nematode of ruminants. Compound 17, known as 6-methoxycomaparvin 5,8-dimethyl ether, showed an inhibitory effect on larval motility (IC50 = 30 μM) and development (IC50 = 31 μM) and induced the eviscerated (Evi) phenotype.
Collapse
Affiliation(s)
- Kah Yean Lum
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Aya C Taki
- Department of Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Robin B Gasser
- Department of Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Ian Tietjen
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Merrick G Ekins
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
- Biodiversity and Geosciences, Queensland Museum, South Brisbane BC, QLD 4101, Australia
| | - Jonathan M White
- School of Chemistry and Bio21 Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Russell S Addison
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Sasha Hayes
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - James St John
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, QLD 4222, Australia
| | - Rohan A Davis
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| |
Collapse
|
3
|
Wang Y, Wang X, Xiong Y, Kaushik AC, Muhammad J, Khan A, Dai H, Wei DQ. New strategy for identifying potential natural HIV-1 non-nucleoside reverse transcriptase inhibitors against drug-resistance: an in silico study. J Biomol Struct Dyn 2019; 38:3327-3341. [PMID: 31422767 DOI: 10.1080/07391102.2019.1656673] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Non-nucleosides reverse transcriptase inhibitors (NNRTIs), specifically targeting the HIV-1 reverse transcriptase (RT), play a unique role in anti-AIDS agents due to their high antiviral potency, structural diversity, and low toxicity in antiretroviral combination therapies used to treat HIV. However, due to the emergence of new drug-resistant strains, the development of novel NNRTIs with adequate potency, improved resistance profiles and less toxicity is highly required. In this work, a novel virtual screening strategy combined with structure-based drug design was proposed to discover the potential inhibitors against drug-resistant HIV strains. Seven structure-variant RTs, ranging from the wild type to a hypothetical multi-mutant were regarded as target proteins to perform structure-based virtual screening. Totally 23 small molecules with good binding affinity were identified from the Traditional Chinese Medicine database (TCM) as potential NNRTIs candidates. Among these hits, (+)-Hinokinin has confirmed anti-HIV activity, and some hits are structurally identical with anti-HIV compounds. Almost all these hits are consistent with external experimental results. Molecular simulations analysis revealed that top 2 hits (Pallidisetin A and Pallidisetin B) bind stably and in high affinity to HIV-RT, which are ready to be experimental confirmed. These results suggested that the strategy we proposed is feasible, trustworthy and effective. Our finding might be helpful in the identification of novel NNRTIs against drug-resistant, and also provide a new clue for the discovery of HIV drugs in natural products.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yanjing Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.,Peng Cheng Laboratory, Nanshan District, Shenzhen, Guangdong, China
| | - Xiangeng Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.,Peng Cheng Laboratory, Nanshan District, Shenzhen, Guangdong, China
| | - Yi Xiong
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Aman Chandra Kaushik
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Junaid Muhammad
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Abbas Khan
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Dai
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, and Joint Laboratory of International Cooperation in Metabolic and Developmental Sciences, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
4
|
Chromenone derivatives as a versatile scaffold with dual mode of inhibition of HIV-1 reverse transcriptase-associated Ribonuclease H function and integrase activity. Eur J Med Chem 2019; 182:111617. [PMID: 31442684 DOI: 10.1016/j.ejmech.2019.111617] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/09/2019] [Accepted: 08/10/2019] [Indexed: 12/31/2022]
Abstract
A number of compounds targeting different processes of the Human Immunodeficiency Virus type 1 (HIV-1) life cycle have been developed in the continuing fight against AIDS. Coumarin-based molecules already proved to act as HIV-1 Protease (PR) or Integrase (IN) inhibitors and also to target HIV-1 reverse transcriptase (RT), blocking the DNA-dependent DNA-polymerase activity or the RNA-dependent DNA-polymerase activity working as common NNRTIs. In the present study, with the aim to exploit a coumarin-based scaffold to achieve the inhibition of multiple viral coded enzymatic functions, novel 4-hydroxy-2H, 5H-pyrano (3, 2-c) chromene-2, 5-dione derivatives were synthesized. The modeling studies calculated the theoretical binding affinity of the synthesized compounds on both HIV-1 IN and RT-associated Ribonuclease H (RNase H) active sites, which was confirmed by biological assays. Our results provide a basis for the identification of dual HIV-1 IN and RT RNase H inhibitors compounds.
Collapse
|
5
|
Feilcke R, Arnouk G, Raphane B, Richard K, Tietjen I, Andrae-Marobela K, Erdmann F, Schipper S, Becker K, Arnold N, Frolov A, Reiling N, Imming P, Fobofou SAT. Biological activity and stability analyses of knipholone anthrone, a phenyl anthraquinone derivative isolated from Kniphofia foliosa Hochst. J Pharm Biomed Anal 2019; 174:277-285. [PMID: 31185339 DOI: 10.1016/j.jpba.2019.05.065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/13/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Abstract
Knipholone (1) and knipholone anthrone (2), isolated from the Ethiopian medicinal plant Kniphofia foliosa Hochst. are two phenyl anthraquinone derivatives, a compound class known for biological activity. In the present study, we describe the activity of both 1 and 2 in several biological assays including cytotoxicity against four human cell lines (Jurkat, HEK293, SH-SY5Y and HT-29), antiplasmodial activity against Plasmodium falciparum 3D7 strain, anthelmintic activity against the model organism Caenorhabditis elegans, antibacterial activity against Aliivibrio fischeri and Mycobacterium tuberculosis and anti-HIV-1 activity in peripheral blood mononuclear cells (PBMCs) infected with HIV-1c. In parallel, we investigated the stability of knipholone (2) in solution and in culture media. Compound 1 displays strong cytotoxicity against Jurkat, HEK293 and SH-SY5Y cells with growth inhibition ranging from approximately 62-95% when added to cells at 50 μM, whereas KA (2) exhibits weak to strong activity with 26, 48 and 70% inhibition of cell growth, respectively. Both 1 and 2 possess significant antiplasmodial activity against Plasmodium falciparum 3D7 strain with IC50 values of 1.9 and 0.7 μM, respectively. These results complement previously reported data on the cytotoxicity and antiplasmodial activity of 1 and 2. Furthermore, compound 2 showed HIV-1c replication inhibition (growth inhibition higher than 60% at tested concentrations 0.5, 5, 15 and 50 μg/ml and an EC50 value of 4.3 μM) associated with cytotoxicity against uninfected PBMCs. The stability study based on preincubation, HPLC and APCI-MS (atmospheric-pressure chemical ionization mass spectrometry) analysis indicates that compound 2 is unstable in culture media and readily oxidizes to form compound 1. Therefore, the biological activity attributed to 2 might be influenced by its degradation products in media including 1 and other possible dimers. Hence, bioactivity results previously reported from this compound should be taken with caution and checked if they differ from those of its degradation products. To the best of our knowledge, this is the first report on the anti-HIV activity and stability analysis of compound 2.
Collapse
Affiliation(s)
- Ruth Feilcke
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany
| | - Georgette Arnouk
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany
| | - Boingotlo Raphane
- Department of Biological Sciences, Faculty of Science, University of Botswana, Block 235, Private Bag, 0022 Gaborone, Botswana
| | - Khumoekae Richard
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Ian Tietjen
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Kerstin Andrae-Marobela
- Department of Biological Sciences, Faculty of Science, University of Botswana, Block 235, Private Bag, 0022 Gaborone, Botswana
| | - Frank Erdmann
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany
| | - Susanne Schipper
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany
| | - Katja Becker
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany
| | - Norbert Arnold
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany
| | - Andrej Frolov
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany; Department of Biochemistry, St. Petersburg State University, 199904 St. Petersburg, Russia
| | - Norbert Reiling
- Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel, 23845 Borstel, Germany
| | - Peter Imming
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany
| | - Serge A T Fobofou
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle (Saale), Germany; Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany.
| |
Collapse
|
6
|
Di Petrillo A, Fais A, Pintus F, Santos-Buelga C, González-Paramás AM, Piras V, Orrù G, Mameli A, Tramontano E, Frau A. Broad-range potential of Asphodelus microcarpus leaves extract for drug development. BMC Microbiol 2017; 17:159. [PMID: 28709400 PMCID: PMC5513112 DOI: 10.1186/s12866-017-1068-5] [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: 06/01/2017] [Accepted: 07/10/2017] [Indexed: 02/07/2023] Open
Abstract
Background Many plants have been used in traditional medicine for their antibacterial, antifungal, antiprotozoal, antiviral, antidiarrhoeal, analgesic, antimalarial, antioxidant, anti-inflammatory and anticancer activities. In order to find novel antimicrobial and antiviral agents, the aim of the present study was the evaluation of the antibacterial and antibiofilm susceptibility of Asphodelus microcarpus leaves extract. Moreover, the antiviral activity and the phytochemical composition of the active extract were also determined. Methods Antimicrobial and antibiofilm activities of leaves ethanol extract of A. microcarpus were evaluated on 13 different microbial strains. We selected three different sets of microorganisms: (i) Gram-positive bacteria, (ii) Gram-negative bacteria and (iii) yeasts. The potential antiviral activity of A. microcarpus leaves ethanol extract was evaluated with a luciferase reporter gene assay in which the dsRNA-dependent RIG-I-mediated IFN-β activation was inducted or inhibited by the Ebola virus VP35 protein. HPLC-DAD-MS was used to identify phenolic profile of the active extract. Results A. microcarpus leaves extract showed a potent inhibitory activity on Gram-positive bacteria while only a reduced inhibition was observed on Gram-negative bacteria. No activity was detected against Yeasts. The extract also showed an interesting antibiofilm motif on various bacterial strains (E. coli, S. aureus, S. haemolyticus and B. clausii). Moreover, this extract significantly affected the Ebola virus VP35 inhibition of the viral RNA (vRNA) induced IFN response. Conclusions The overall results provide supportive data on the use of A. microcarpus as antimicrobial agent and a potential source of anti-viral natural products. Data collected set the bases for further studies for the identification of single active components and the development of new pharmaceuticals.
Collapse
Affiliation(s)
- Amalia Di Petrillo
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, SS 554, Bivio per Sestu, I-09042, Cagliari, Monserrato, Italy
| | - Antonella Fais
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, SS 554, Bivio per Sestu, I-09042, Cagliari, Monserrato, Italy.
| | - Francesca Pintus
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, SS 554, Bivio per Sestu, I-09042, Cagliari, Monserrato, Italy
| | - Celestino Santos-Buelga
- Grupo de Investigación en Polifenoles, Unidad de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Salamanca, Salamanca, Spain
| | - Ana M González-Paramás
- Grupo de Investigación en Polifenoles, Unidad de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Salamanca, Salamanca, Spain
| | - Vincenzo Piras
- Department of Surgical Science, University of Cagliari, 09124, Cagliari, Italy
| | - Germano Orrù
- Department of Surgical Science, University of Cagliari, 09124, Cagliari, Italy
| | - Antonello Mameli
- Department of Surgical Science, University of Cagliari, 09124, Cagliari, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, SS 554, Bivio per Sestu, I-09042, Cagliari, Monserrato, Italy
| | - Aldo Frau
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, SS 554, Bivio per Sestu, I-09042, Cagliari, Monserrato, Italy
| |
Collapse
|
7
|
Martini R, Esposito F, Corona A, Ferrarese R, Ceresola ER, Visconti L, Tintori C, Barbieri A, Calcaterra A, Iovine V, Canducci F, Tramontano E, Botta M. Natural Product Kuwanon-L Inhibits HIV-1 Replication through Multiple Target Binding. Chembiochem 2017; 18:374-377. [PMID: 27992102 DOI: 10.1002/cbic.201600592] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Indexed: 12/30/2022]
Abstract
In recent years many advances have been made in the fight against HIV-1 infection. However, the lack of a vaccine, together with the increasing resistance to the highly active anti-retroviral therapy (HAART), make HIV-1 infection still a serious global emergency. Thus, new compounds with original modes of action are continuously required, and natural products have ever been a very interesting class of pharmacologically active molecules. Some of them have been used since ancient times against viral infections. Here we present a work in which we suggest that kuwanon-L, a natural product active as an HIV-1 integrase (IN) inhibitor, might exert its overall antiviral activity through binding to multiple viral targets. Specific enzymatic tests, together with a time-of-addition (TOA) experiment, support our hypothesis of binding both to IN and to reverse transcriptase (RT). Overall, this compound can be considered an attractive lead for the development of new classes of antiviral agents able to overcome the problem of resistance, due to its ability to exert its action by binding simultaneously to multiple viral targets.
Collapse
Affiliation(s)
- Riccardo Martini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Francesca Esposito
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, SS 554, 09042, Monserrato, Cagliari, Italy
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, SS 554, 09042, Monserrato, Cagliari, Italy
| | - Roberto Ferrarese
- Laboratory of Microbiology, San Raffaele Hospital, IRCCS, Via Olgettina 58, 20132, Milan, Italy
| | - Elisa Rita Ceresola
- Department of Biotechnology and Life Sciences, University of Insubria, Via Ravasi 2, 21100, Varese, Italy
| | - Laura Visconti
- Laboratory of Microbiology, San Raffaele Hospital, IRCCS, Via Olgettina 58, 20132, Milan, Italy
| | - Cristina Tintori
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Alessandro Barbieri
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Andrea Calcaterra
- Department of Chemistry and Technology of Drugs, University of Rome "Sapienza", Piazzale Aldo Moro 5, 00185, Roma, Italy
| | - Valentina Iovine
- Department of Chemistry and Technology of Drugs, University of Rome "Sapienza", Piazzale Aldo Moro 5, 00185, Roma, Italy
| | - Filippo Canducci
- Laboratory of Microbiology, San Raffaele Hospital, IRCCS, Via Olgettina 58, 20132, Milan, Italy.,Department of Biotechnology and Life Sciences, University of Insubria, Via Ravasi 2, 21100, Varese, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, SS 554, 09042, Monserrato, Cagliari, Italy
| | - Maurizio Botta
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.,Lead Discovery Siena s.r.l., Via Vittorio Alfieri 31, 53019, Castelnuovo Berardenga, Italy
| |
Collapse
|
8
|
Esposito F, Carli I, Del Vecchio C, Xu L, Corona A, Grandi N, Piano D, Maccioni E, Distinto S, Parolin C, Tramontano E. Sennoside A, derived from the traditional chinese medicine plant Rheum L., is a new dual HIV-1 inhibitor effective on HIV-1 replication. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1383-1391. [PMID: 27765358 DOI: 10.1016/j.phymed.2016.08.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/19/2016] [Accepted: 08/09/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Despite the availability of effective antiretroviral therapies, drugs for HIV-1 treatment with new mode of action are still needed. An innovative approach is aimed to identify dual HIV-1 inhibitors, small molecules that can inhibit two viral functions at the same time. Rhubarb, originated from Rheum palmatum L. and Rheum officinale Baill., is one of the earliest and most commonly used medicinal plants in Traditional Chinese Medicine (TCM) practice. We wanted to explore TCM for the identification of new chemical scaffolds with dual action abilities against HIV-1. METHODS R. palmatum L. and R. officinale Baill. extracts along with their main single isolated constituents anthraquinone derivatives were tested on both HIV-1 Reverse Transcriptase (RT)-associated DNA Polymerase (RDDP) and Ribonuclease H (RNase H) activities in biochemical assays. Active compounds were then assayed for their effects on HIV-1 mutated RTs, integrase (IN) and viral replication. RESULTS Both R. palmatum L. and R. officinale Baill. extracts inhibited the HIV-1 RT-associated RNase H activity. Among the isolated constituents, Sennoside A and B were effective on both RDDP and RNase H RT-associated functions in biochemical assays. Sennoside A was less potent when tested on K103N, Y181C, Y188L, N474A and Q475A mutated RTs, suggesting the involvement of two RT binding sites for its antiviral activity. Sennoside A affected also HIV-1 IN activity in vitro and HIV-1 replication in cell-based assays. Viral DNA production and time of addition studies showed that Sennoside A targets the HIV-1 reverse transcription process. CONCLUSION Sennoside A is a new scaffold for the development of HIV-1 dual RT inhibitors.
Collapse
Affiliation(s)
- Francesca Esposito
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella di Monserrato SS554, 09042, Monserrato, Cagliari, Italy
| | - Ilaria Carli
- Department of Molecular Medicine, University of Padova, via Gabelli 63, 35121 Padova, Italy
| | - Claudia Del Vecchio
- Department of Molecular Medicine, University of Padova, via Gabelli 63, 35121 Padova, Italy
| | - Lijia Xu
- Institute of Medicinal Plant Development (IMPLAD), 151 Malianwa North Road Haidian District, 100193 Beijing, China
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella di Monserrato SS554, 09042, Monserrato, Cagliari, Italy
| | - Nicole Grandi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella di Monserrato SS554, 09042, Monserrato, Cagliari, Italy
| | - Dario Piano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella di Monserrato SS554, 09042, Monserrato, Cagliari, Italy
| | - Elias Maccioni
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella di Monserrato SS554, 09042, Monserrato, Cagliari, Italy
| | - Simona Distinto
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella di Monserrato SS554, 09042, Monserrato, Cagliari, Italy
| | - Cristina Parolin
- Department of Molecular Medicine, University of Padova, via Gabelli 63, 35121 Padova, Italy.
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella di Monserrato SS554, 09042, Monserrato, Cagliari, Italy; Genetics and Biomedical Research institute, National Research Council (CNR), Cittadella di Monserrato SS554, 09042, Monserrato, Cagliari, Italy.
| |
Collapse
|
9
|
Pala N, Esposito F, Rogolino D, Carcelli M, Sanna V, Palomba M, Naesens L, Corona A, Grandi N, Tramontano E, Sechi M. Inhibitory Effect of 2,3,5,6-Tetrafluoro-4-[4-(aryl)-1H-1,2,3-triazol-1-yl]benzenesulfonamide Derivatives on HIV Reverse Transcriptase Associated RNase H Activities. Int J Mol Sci 2016; 17:E1371. [PMID: 27556447 PMCID: PMC5000766 DOI: 10.3390/ijms17081371] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/12/2016] [Accepted: 08/15/2016] [Indexed: 12/23/2022] Open
Abstract
The HIV-1 ribonuclease H (RNase H) function of the reverse transcriptase (RT) enzyme catalyzes the selective hydrolysis of the RNA strand of the RNA:DNA heteroduplex replication intermediate, and represents a suitable target for drug development. A particularly attractive approach is constituted by the interference with the RNase H metal-dependent catalytic activity, which resides in the active site located at the C-terminus p66 subunit of RT. Herein, we report results of an in-house screening campaign that allowed us to identify 4-[4-(aryl)-1H-1,2,3-triazol-1-yl]benzenesulfonamides, prepared by the "click chemistry" approach, as novel potential HIV-1 RNase H inhibitors. Three compounds (9d, 10c, and 10d) demonstrated a selective inhibitory activity against the HIV-1 RNase H enzyme at micromolar concentrations. Drug-likeness, predicted by the calculation of a panel of physicochemical and ADME properties, putative binding modes for the active compounds, assessed by computational molecular docking, as well as a mechanistic hypothesis for this novel chemotype are reported.
Collapse
Affiliation(s)
- Nicolino Pala
- Dipartimento di Chimica e Farmacia, Università di Sassari, Via Vienna 2, I-07100 Sassari, Italy.
| | - Francesca Esposito
- Dipartimento di Scienze della Vita e dell'Ambiente-Sezione Biomedica, Università di Cagliari, Cittadella Universitaria SS554, I-09042 Monserrato, Italy.
| | - Dominga Rogolino
- Dipartimento di Chimica, Università di Parma, Parco Area delle Scienze 17/A, I-43124 Parma, Italy.
| | - Mauro Carcelli
- Dipartimento di Chimica, Università di Parma, Parco Area delle Scienze 17/A, I-43124 Parma, Italy.
| | - Vanna Sanna
- Dipartimento di Chimica e Farmacia, Università di Sassari, Via Vienna 2, I-07100 Sassari, Italy.
| | - Michele Palomba
- Dipartimento di Chimica e Farmacia, Università di Sassari, Via Vienna 2, I-07100 Sassari, Italy.
| | - Lieve Naesens
- Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium.
| | - Angela Corona
- Dipartimento di Scienze della Vita e dell'Ambiente-Sezione Biomedica, Università di Cagliari, Cittadella Universitaria SS554, I-09042 Monserrato, Italy.
| | - Nicole Grandi
- Dipartimento di Scienze della Vita e dell'Ambiente-Sezione Biomedica, Università di Cagliari, Cittadella Universitaria SS554, I-09042 Monserrato, Italy.
| | - Enzo Tramontano
- Dipartimento di Scienze della Vita e dell'Ambiente-Sezione Biomedica, Università di Cagliari, Cittadella Universitaria SS554, I-09042 Monserrato, Italy.
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), I-09042 Monserrato, Italy.
| | - Mario Sechi
- Dipartimento di Chimica e Farmacia, Università di Sassari, Via Vienna 2, I-07100 Sassari, Italy.
| |
Collapse
|
10
|
Hunegnaw R, Vassylyeva M, Dubrovsky L, Pushkarsky T, Sviridov D, Anashkina AA, Üren A, Brichacek B, Vassylyev DG, Adzhubei AA, Bukrinsky M. Interaction Between HIV-1 Nef and Calnexin: From Modeling to Small Molecule Inhibitors Reversing HIV-Induced Lipid Accumulation. Arterioscler Thromb Vasc Biol 2016; 36:1758-71. [PMID: 27470515 DOI: 10.1161/atvbaha.116.307997] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 07/13/2016] [Indexed: 01/22/2023]
Abstract
OBJECTIVE HIV-infected patients are at an increased risk of developing atherosclerosis, in part because of downmodulation and functional impairment of ATP-binding cassette A1 (ABCA1) cholesterol transporter by the HIV-1 protein Nef. The mechanism of this effect involves Nef interacting with an ER chaperone calnexin and disrupting calnexin binding to ABCA1, leading to ABCA1 retention in ER, its degradation and resulting suppression of cholesterol efflux. However, molecular details of Nef-calnexin interaction remained unknown, limiting the translational impact of this finding. APPROACH AND RESULTS Here, we used molecular modeling and mutagenesis to characterize Nef-calnexin interaction and to identify small molecule compounds that could block it. We demonstrated that the interaction between Nef and calnexin is direct and can be reconstituted using recombinant proteins in vitro with a binding affinity of 89.1 nmol/L measured by surface plasmon resonance. The cytoplasmic tail of calnexin is essential and sufficient for interaction with Nef, and binds Nef with an affinity of 9.4 nmol/L. Replacing lysine residues in positions 4 and 7 of Nef with alanines abrogates Nef-calnexin interaction, prevents ABCA1 downregulation by Nef, and preserves cholesterol efflux from HIV-infected cells. Through virtual screening of the National Cancer Institute library of compounds, we identified a compound, 1[(7-oxo-7H-benz[de]anthracene-3-yl)amino]anthraquinone, which blocked Nef-calnexin interaction, partially restored ABCA1 activity in HIV-infected cells, and reduced foam cell formation in a culture of HIV-infected macrophages. CONCLUSION This study identifies potential targets that can be exploited to block the pathogenic effect of HIV infection on cholesterol metabolism and prevent atherosclerosis in HIV-infected subjects.
Collapse
Affiliation(s)
- Ruth Hunegnaw
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Marina Vassylyeva
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Larisa Dubrovsky
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Tatiana Pushkarsky
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Dmitri Sviridov
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Anastasia A Anashkina
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Aykut Üren
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Beda Brichacek
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Dmitry G Vassylyev
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Alexei A Adzhubei
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü).
| | - Michael Bukrinsky
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü).
| |
Collapse
|
11
|
Biological Activities of Aerial Parts Extracts of Euphorbia characias. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1538703. [PMID: 27314007 PMCID: PMC4895043 DOI: 10.1155/2016/1538703] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/27/2016] [Accepted: 05/03/2016] [Indexed: 11/17/2022]
Abstract
The aim of the present study was to evaluate antioxidant, antimicrobial, anti-HIV, and cholinesterase inhibitory activities of aqueous and alcoholic extracts from leaves, stems, and flowers of Euphorbia characias. The extracts showed a high antioxidant activity and were a good source of total polyphenols and flavonoids. Ethanolic extracts from leaves and flowers displayed the highest inhibitory activity against acetylcholinesterase and butyrylcholinesterase, showing potential properties against Alzheimer's disease. Antimicrobial assay showed that leaves and flowers extracts were active against all Gram-positive bacteria tested. The ethanolic leaves extract appeared to have the strongest antibacterial activity against Bacillus cereus with MIC value of 312.5 μg/mL followed by Listeria monocytogenes and Staphylococcus aureus that also exhibited good sensitivity with MIC values of 1250 μg/mL. Moreover, all the extracts possessed anti-HIV activity. The ethanolic flower extract was the most potent inhibitor of HIV-1 RT DNA polymerase RNA-dependent and Ribonuclease H with IC50 values of 0.26 and 0.33 μg/mL, respectively. The LC-DAD metabolic profile showed that ethanolic leaves extract contains high levels of quercetin derivatives. This study suggests that Euphorbia characias extracts represent a good source of natural bioactive compounds which could be useful for pharmaceutical application as well as in food system for the prevention of the growth of food-borne bacteria and to extend the shelf-life of processed foods.
Collapse
|
12
|
Corona A, Desantis J, Massari S, Distinto S, Masaoka T, Sabatini S, Esposito F, Manfroni G, Maccioni E, Cecchetti V, Pannecouque C, Le Grice SFJ, Tramontano E, Tabarrini O. Studies on Cycloheptathiophene-3-carboxamide Derivatives as Allosteric HIV-1 Ribonuclease H Inhibitors. ChemMedChem 2016; 11:1709-20. [PMID: 26990134 DOI: 10.1002/cmdc.201600015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Indexed: 02/04/2023]
Abstract
Despite the significant progress achieved with combination antiretroviral therapy in the fight against human immunodeficiency virus (HIV) infection, the difficulty to eradicate the virus together with the rapid emergence of multidrug-resistant strains clearly underline a pressing need for innovative agents, possibly endowed with novel mechanisms of action. In this context, owing to its essential role in HIV genome replication, the reverse transcriptase associated ribonuclease H (RNase H) has proven to be an appealing target. To identify new RNase H inhibitors, an in-house cycloheptathiophene-3-carboxamide library was screened; this led to compounds endowed with inhibitory activity, the structural optimization of which led to the catechol derivative 2-(3,4-dihydroxybenzamido)-N-(pyridin-2-yl)-5,6,7,8-tetrahydro-4H-cyclohepta[b]thiophene-3-carboxamide (compound 33) with an IC50 value on the RNase H activity in the nanomolar range. Mechanistic studies suggested selective inhibition of the RNase H through binding to an innovative allosteric site, which could be further exploited to enrich this class of inhibitors.
Collapse
Affiliation(s)
- Angela Corona
- Dipartimento di Scienze della Vita e dell'Ambiente, Cittadella Universitaria di Monserrato, SS554, 09042, Monserrato, Italy
| | - Jenny Desantis
- Dipartimento di Scienze Farmaceutiche, Università di Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Serena Massari
- Dipartimento di Scienze Farmaceutiche, Università di Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Simona Distinto
- Dipartimento di Scienze della Vita e dell'Ambiente, Cittadella Universitaria di Monserrato, SS554, 09042, Monserrato, Italy
| | - Takashi Masaoka
- Basic Research Laboratory, National Cancer Institute, Frederick, MD, 21702-1201, USA
| | - Stefano Sabatini
- Dipartimento di Scienze Farmaceutiche, Università di Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Francesca Esposito
- Dipartimento di Scienze della Vita e dell'Ambiente, Cittadella Universitaria di Monserrato, SS554, 09042, Monserrato, Italy
| | - Giuseppe Manfroni
- Dipartimento di Scienze Farmaceutiche, Università di Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Elias Maccioni
- Dipartimento di Scienze della Vita e dell'Ambiente, Cittadella Universitaria di Monserrato, SS554, 09042, Monserrato, Italy
| | - Violetta Cecchetti
- Dipartimento di Scienze Farmaceutiche, Università di Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Christophe Pannecouque
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research-KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| | - Stuart F J Le Grice
- Basic Research Laboratory, National Cancer Institute, Frederick, MD, 21702-1201, USA
| | - Enzo Tramontano
- Dipartimento di Scienze della Vita e dell'Ambiente, Cittadella Universitaria di Monserrato, SS554, 09042, Monserrato, Italy.
| | - Oriana Tabarrini
- Dipartimento di Scienze Farmaceutiche, Università di Perugia, Via del Liceo 1, 06123, Perugia, Italy.
| |
Collapse
|
13
|
Schneider A, Corona A, Spöring I, Jordan M, Buchholz B, Maccioni E, Di Santo R, Bodem J, Tramontano E, Wöhrl BM. Biochemical characterization of a multi-drug resistant HIV-1 subtype AG reverse transcriptase: antagonism of AZT discrimination and excision pathways and sensitivity to RNase H inhibitors. Nucleic Acids Res 2016; 44:2310-22. [PMID: 26850643 PMCID: PMC4797301 DOI: 10.1093/nar/gkw060] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 01/24/2016] [Indexed: 11/27/2022] Open
Abstract
We analyzed a multi-drug resistant (MR) HIV-1 reverse transcriptase (RT), subcloned from a patient-derived subtype CRF02_AG, harboring 45 amino acid exchanges, amongst them four thymidine analog mutations (TAMs) relevant for high-level AZT (azidothymidine) resistance by AZTMP excision (M41L, D67N, T215Y, K219E) as well as four substitutions of the AZTTP discrimination pathway (A62V, V75I, F116Y and Q151M). In addition, K65R, known to antagonize AZTMP excision in HIV-1 subtype B was present. Although MR-RT harbored the most significant amino acid exchanges T215Y and Q151M of each pathway, it exclusively used AZTTP discrimination, indicating that the two mechanisms are mutually exclusive and that the Q151M pathway is obviously preferred since it confers resistance to most nucleoside inhibitors. A derivative was created, additionally harboring the TAM K70R and the reversions M151Q as well as R65K since K65R antagonizes excision. MR-R65K-K70R-M151Q was competent of AZTMP excision, whereas other combinations thereof with only one or two exchanges still promoted discrimination. To tackle the multi-drug resistance problem, we tested if the MR-RTs could still be inhibited by RNase H inhibitors. All MR-RTs exhibited similar sensitivity toward RNase H inhibitors belonging to different inhibitor classes, indicating the importance of developing RNase H inhibitors further as anti-HIV drugs.
Collapse
Affiliation(s)
- Anna Schneider
- Universität Bayreuth, Lehrstuhl Biopolymere und Forschungszentrum für Bio-Makromoleküle, Universitätsstrasse 30, 95447 Bayreuth, Germany
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, SS 554, 09042, Monserrato, Cagliari, Italy
| | - Imke Spöring
- Julius-Maximilians-Universität Würzburg, Institut für Virologie und Immunbiologie, Versbacher Strasse 7, 97078 Würzburg, Germany
| | - Mareike Jordan
- Universität Bayreuth, Lehrstuhl Biopolymere und Forschungszentrum für Bio-Makromoleküle, Universitätsstrasse 30, 95447 Bayreuth, Germany
| | - Bernd Buchholz
- Universität Heidelberg, Medizinische Fakultät Mannheim, Klinik für Kinder- und Jugendmedizin, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Elias Maccioni
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, SS 554, 09042, Monserrato, Cagliari, Italy
| | - Roberto Di Santo
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, "Sapienza" Università di Roma, Rome, I-00185, Italy
| | - Jochen Bodem
- Julius-Maximilians-Universität Würzburg, Institut für Virologie und Immunbiologie, Versbacher Strasse 7, 97078 Würzburg, Germany
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, SS 554, 09042, Monserrato, Cagliari, Italy
| | - Birgitta M Wöhrl
- Universität Bayreuth, Lehrstuhl Biopolymere und Forschungszentrum für Bio-Makromoleküle, Universitätsstrasse 30, 95447 Bayreuth, Germany
| |
Collapse
|
14
|
Chingwaru W, Vidmar J, Kapewangolo PT. The Potential of Sub-Saharan African Plants in the Management of Human Immunodeficiency Virus Infections: A Review. Phytother Res 2015; 29:1452-87. [PMID: 26337608 DOI: 10.1002/ptr.5433] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 07/09/2015] [Accepted: 07/22/2015] [Indexed: 12/28/2022]
Abstract
Acquired immunodeficiency syndrome, caused by human immunodeficiency virus (HIV), is a leading cause of mortality and morbidity in Sub-Saharan Africa, particularly in Southern Africa. Phytomedicines are an integral part of African health care. The Southern African flora is composed of at least 23 400 taxa. Despite this richness, only a handful of botanical products have been assessed for activities against HIV. This study aimed to summarize the potential of Sub-Saharan African plants, based on their composition and the established bioactivities, as sources of agents to manage HIV symptoms and as retroviral therapy. At least 109 plant species from 42 families and 94 genera that are found in Southern Africa were shown to have potential or actual activities against HIV. Only 12 of these plant species from 6 families and 10 genera were shown to harbour anti-HIV properties. Phytochemicals that include β-sitosterols, terpenoids, glycosides, saponins, flavonoids, triterpenoids, tannins and alkaloids, which harbour anti-HIV properties, were found to have a near cosmopolitan presence across the plant families in the region. Bioactivities of multiple phytochemicals are comparable to those for standard allopathic antiretroviral drugs. Research to determine the anti-HIV activities of the identified and other plants, including clinical trials, is long overdue.
Collapse
Affiliation(s)
- Walter Chingwaru
- Department of Biological Sciences, Faculty of Science, Bindura University of Science Education, P. Bag 1020, Bindura, Zimbabwe.,Institute Ceres/Zavod Ceres, Lahovna 16, 3000, Celje, Slovenia
| | - Jerneja Vidmar
- Institute Ceres/Zavod Ceres, Lahovna 16, 3000, Celje, Slovenia.,Department of Plastic and Reconstructive Surgery, University Medical Centre Maribor, Ljubljanska 5, 2000, Maribor, Slovenia
| | - Petrina T Kapewangolo
- Department of Chemistry and Biochemistry, University of Namibia, P/Bag 13301, 340 Mandume Ndemufayo Avenue, Pionierspark, Windhoek, Namibia
| |
Collapse
|
15
|
Cuzzucoli Crucitti G, Métifiot M, Pescatori L, Messore A, Madia VN, Pupo G, Saccoliti F, Scipione L, Tortorella S, Esposito F, Corona A, Cadeddu M, Marchand C, Pommier Y, Tramontano E, Costi R, Di Santo R. Structure-activity relationship of pyrrolyl diketo acid derivatives as dual inhibitors of HIV-1 integrase and reverse transcriptase ribonuclease H domain. J Med Chem 2015; 58:1915-28. [PMID: 25629256 DOI: 10.1021/jm501799k] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The development of HIV-1 dual inhibitors is a highly innovative approach aimed at reducing drug toxic side effects as well as therapeutic costs. HIV-1 integrase (IN) and reverse transcriptase-associated ribonuclease H (RNase H) are both selective targets for HIV-1 chemotherapy, and the identification of dual IN/RNase H inhibitors is an attractive strategy for new drug development. We newly synthesized pyrrolyl derivatives that exhibited good potency against IN and a moderate inhibition of the RNase H function of RT, confirming the possibility of developing dual HIV-1 IN/RNase H inhibitors and obtaining new information for the further development of more effective dual HIV-1 inhibitors.
Collapse
Affiliation(s)
- Giuliana Cuzzucoli Crucitti
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, "Sapienza" Università di Roma , Rome, I-00185, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Inhibition of the DNA polymerase and RNase H activities of HIV-1 reverse transcriptase and HIV-1 replication by Brasenia schreberi (Junsai) and Petasites japonicus (Fuki) components. J Nat Med 2015; 69:432-40. [PMID: 25663480 DOI: 10.1007/s11418-015-0885-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/31/2014] [Indexed: 01/22/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) possesses two distinct enzymatic activities: those of RNA- and DNA-dependent DNA polymerases and RNase H. In the current HIV-1 therapy, all HIV-1 RT inhibitors inhibit the activity of DNA polymerase, but not that of RNase H. We previously reported that ethanol and water extracts of Brasenia schreberi (Junsai) inhibited the DNA polymerase activity of HIV-1 RT [Hisayoshi et al. (2014) J Biol Macromol 14:59-65]. In this study, we screened 43 edible plants and found that ethanol and water extracts of Brasenia schreberi and water extract of Petasites japonicus strongly inhibit not only the activity of DNA polymerase to incorporate dTTP into poly(rA)-p(dT)15 but also the activity of RNase H to hydrolyze the RNA strand of an RNA/DNA hybrid. In addition, these three extracts inhibit HIV-1 replication in human cells, with EC50 values of 1-2 µg/ml. These results suggest that Brasenia schreberi and Petasites japonicus contain substances that block HIV-1 replication by inhibiting the DNA polymerase activity and/or RNase H activity of HIV-1 RT.
Collapse
|
17
|
Alam Z, Al-Mahdi Z, Zhu Y, McKee Z, Parris DS, Parikh HI, Kellogg GE, Kuchta A, McVoy MA. Anti-cytomegalovirus activity of the anthraquinone atanyl blue PRL. Antiviral Res 2014; 114:86-95. [PMID: 25499125 PMCID: PMC4289655 DOI: 10.1016/j.antiviral.2014.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 11/28/2022]
Abstract
The anthraquinone atanyl blue PRL inhibits human cytomegalovirus replication. The block to viral replication appears early after entry and substantially reduces viral immediate early gene expression. In vitro, atanyl blue PRL inhibits the nuclease activity of purified viral alkaline nuclease, UL98. The antiviral activity of atanyl blue PRL may be manifested through inhibition of UL98’s nuclease activity.
Human cytomegalovirus (CMV) causes significant disease in immunocompromised patients and serious birth defects if acquired in utero. Available CMV antivirals target the viral DNA polymerase, have significant toxicities, and suffer from resistance. New drugs targeting different pathways would be beneficial. The anthraquinone emodin is proposed to inhibit herpes simplex virus by blocking the viral nuclease. Emodin and related anthraquinones are also reported to inhibit CMV. In the present study, emodin reduced CMV infectious yield with an EC50 of 4.9 μM but was cytotoxic at concentrations only twofold higher. Related anthraquinones acid blue 40 and alizarin violet R inhibited CMV at only high concentrations (238–265 μM) that were also cytotoxic. However, atanyl blue PRL inhibited infectious yield of CMV with an EC50 of 6.3 μM, significantly below its 50% cytotoxic concentration of 216 μM. Atanyl blue PRL reduced CMV infectivity and inhibited spread. When added up to 1 h after infection, it dramatically reduced CMV immediate early protein expression and blocked viral DNA synthesis. However, it had no antiviral activity when added 24 h after infection. Interestingly, atanyl blue PRL inhibited nuclease activities of purified CMV UL98 protein with IC50 of 4.5 and 9.3 μM. These results indicate that atanyl blue PRL targets very early post-entry events in CMV replication and suggest it may act through inhibition of UL98, making it a novel CMV inhibitor. This compound may provide valuable insights into molecular events that occur at the earliest times post-infection and serve as a lead structure for antiviral development.
Collapse
Affiliation(s)
- Zohaib Alam
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Zainab Al-Mahdi
- Medical Science Department, College of Nursing, University of Babylon, Babylon, Iraq
| | - Yali Zhu
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Zachary McKee
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Deborah S Parris
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Hardik I Parikh
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Glen E Kellogg
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Alison Kuchta
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Michael A McVoy
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA.
| |
Collapse
|
18
|
Meleddu R, Cannas V, Distinto S, Sarais G, Del Vecchio C, Esposito F, Bianco G, Corona A, Cottiglia F, Alcaro S, Parolin C, Artese A, Scalise D, Fresta M, Arridu A, Ortuso F, Maccioni E, Tramontano E. Design, synthesis, and biological evaluation of 1,3-diarylpropenones as dual inhibitors of HIV-1 reverse transcriptase. ChemMedChem 2014; 9:1869-79. [PMID: 24850787 DOI: 10.1002/cmdc.201402015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Indexed: 12/12/2022]
Abstract
A small library of 1,3-diarylpropenones was designed and synthesized as dual inhibitors of both HIV-1 reverse transcriptase (RT) DNA polymerase (DP) and ribonuclease H (RNase H) associated functions. Compounds were assayed on these enzyme activities, which highlighted dual inhibition properties in the low-micromolar range. Interestingly, mutations in the non-nucleoside RT inhibitor binding pocket strongly affected RNase H inhibition by the propenone derivatives without decreasing their capacity to inhibit DP activity, which suggests long-range RT structural effects. Biochemical and computational studies indicated that the propenone derivatives bind two different interdependent allosteric pockets.
Collapse
Affiliation(s)
- Rita Meleddu
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari (Italy)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Inhibition of foamy virus reverse transcriptase by human immunodeficiency virus type 1 RNase H inhibitors. Antimicrob Agents Chemother 2014; 58:4086-93. [PMID: 24798282 DOI: 10.1128/aac.00056-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
RNase H plays an essential role in the replication of human immunodeficiency virus type 1 (HIV-1). Therefore, it is a promising target for drug development. However, the identification of HIV-1 RNase H inhibitors (RHIs) has been hampered by the open morphology of its active site, the limited number of available RNase H crystal structures in complex with inhibitors, and the fact that, due to the high concentrations of Mg(2+) needed for protein stability, HIV-1 RNase H is not suitable for nuclear magnetic resonance (NMR) inhibitor studies. We recently showed that the RNase H domains of HIV-1 and prototype foamy virus (PFV) reverse transcriptases (RTs) exhibit a high degree of structural similarity. Thus, we examined whether PFV RNase H can serve as an HIV-1 RNase H model for inhibitor interaction studies. Five HIV-1 RHIs inhibited PFV RNase H activity at low-micromolar concentrations similar to those of HIV-1 RNase H, suggesting pocket similarity of the RNase H domains. NMR titration experiments with the PFV RNase H domain and the RHI RDS1643 (6-[1-(4-fluorophenyl)methyl-1H-pyrrol-2-yl)]-2,4-dioxo-5-hexenoic acid ethyl ester) were performed to determine its binding site. Based on these results and previous data, in silico docking analysis showed a putative RDS1643 binding region that reaches into the PFV RNase H active site. Structural overlays were performed with HIV-1 and PFV RNase H to propose the RDS1643 binding site in HIV-1 RNase H. Our results suggest that this approach can be used to establish PFV RNase H as a model system for HIV-1 RNase H in order to identify putative inhibitor binding sites in HIV-1 RNase H.
Collapse
|
20
|
Esposito F, Sanna C, Del Vecchio C, Cannas V, Venditti A, Corona A, Bianco A, Serrilli AM, Guarcini L, Parolin C, Ballero M, Tramontano E. Hypericum hircinum L. components as new single-molecule inhibitors of both HIV-1 reverse transcriptase-associated DNA polymerase and ribonuclease H activities. Pathog Dis 2013; 68:116-24. [PMID: 23821410 DOI: 10.1111/2049-632x.12051] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 03/25/2013] [Accepted: 06/05/2013] [Indexed: 12/01/2022] Open
Abstract
Among HIV-1 reverse transcriptase (RT)-associated functions, DNA polymerase and Ribonuclease H (RNase H) are both essential for HIV replication and excellent targets for drug development. While all RT inhibitors approved for therapy target the DNA polymerase activity, there is the pressing need for new RT inhibitors possibly targeting the RNase H function. In the last 20 years, many natural substances have shown antiviral activity against HIV-1, but only a few against the RNase H function. In this study, we have tested the ethanolic extracts obtained by the Hypericum hircinum L. (Hypericaceae) growing in Sardinia (Italy) on the HIV-1 RT-associated RNase H function and found that they have inhibitory effects. Active extracts were fractionated up to obtain the main components that have been isolated, tested, and identified to be betulinic acid, shikimic acid, chlorogenic acid, quercetin, 5,7,3',5'-tetrahydroxyflavanone, and 5,7,3',5'-tetrahydroxyflavanone 7-O-glucoside. Betulinic acid and 5,7,3',5'-tetrahydroxyflavanone 7-O-glucoside were active on both RT-associated activities, and betulinic acid was also active on HIV-1 mutant RTs resistant to efavirenz. Overall, our results suggest that some of these compounds inhibit the HIV-1 RT binding to an allosteric site previously described for other natural compounds and are potential leads for further drug development of a single molecules having dual inhibitory activity.
Collapse
Affiliation(s)
- Francesca Esposito
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|