1
|
Osella MI, Salazar MO, Solís CM, Furlan RLE. New semisynthetic α-glucosidase inhibitor from a doubly-chemically engineered extract. NATURAL PRODUCTS AND BIOPROSPECTING 2025; 15:4. [PMID: 39755857 DOI: 10.1007/s13659-024-00488-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/03/2024] [Indexed: 01/06/2025]
Abstract
Chemically engineered extracts represent a promising source of new bioactive semi-synthetic molecules. Prepared through direct derivatization of natural extracts, they can include constituents enriched with elements and sub-structures that are less common in natural products compared to drugs. Fourteen such extracts were prepared through sequential reactions with hydrazine and a fluorinating reagent, and their α-glucosidase inhibition properties were compared. For the most bioactive mixture, a chemically modified propolis extract, enzyme inhibition increased 22 times due to the reaction sequence. Bio-guided fractionation led to the isolation of a new fluorinated pyrazole produced within the extract by chemical transformation of the flavonoid chrysin. The inhibitor results from the action of the two reagents used on four common functional groups present in natural products (carbonyl, phenol, aromatic carbon, and a double bond). The reactions led to the opening of a 6-member oxygenated heterocycle to produce a 5-member nitrogenated one, as well as the dehydroxylation and fluorination in two different positions of one of the aromatic rings of the natural starting material, all within a complex mixture of natural products. Overall, these transformations led to an approximately 20-fold increase in the α-glucosidase inhibition by the isolated inhibitor compared to its natural precursor.
Collapse
Affiliation(s)
- María I Osella
- Consejo Nacional de Investigaciones Científicas y Técnicas, Suipacha 531, S2002LRK, Rosario, Argentina
| | - Mario O Salazar
- Consejo Nacional de Investigaciones Científicas y Técnicas, Suipacha 531, S2002LRK, Rosario, Argentina
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK, Rosario, Argentina
| | - Carlos M Solís
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK, Rosario, Argentina
| | - Ricardo L E Furlan
- Consejo Nacional de Investigaciones Científicas y Técnicas, Suipacha 531, S2002LRK, Rosario, Argentina.
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, S2002LRK, Rosario, Argentina.
| |
Collapse
|
2
|
Otsuka H, Nakai K, Shimizu E, Yamaguchi T, Yamano M, Sasaki H, Koyama K, Kinoshita K. Photoreaction products of extract from the fruiting bodies of Polyozellus multiplex. J Nat Med 2024; 78:547-557. [PMID: 38509426 DOI: 10.1007/s11418-024-01790-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 02/12/2024] [Indexed: 03/22/2024]
Abstract
Photochemical reactions are powerful tools for synthesizing organic molecules. The input of energy provided by light offers a means to produce strained and unique molecules that cannot be assembled using thermal protocols, allowing for the production of immense molecular complexity in a single chemical step. Furthermore, unlike thermal reactions, photochemical reactions do not require active reagents such as acids, bases, metals, or enzymes. Photochemical reactions play a central role in green chemistry. This article reports the isolation and structure determination of four new compounds (1-4) from the photoreaction products of the Polyozellus multiplex MeOH ext. The structures of the new compounds were elucidated using MS, IR, comprehensive NMR measurements and microED. The four compounds were formed by deacetylation of polyozellin, the main secondary metabolite of P. multiplex, and addition of singlet oxygen generated by sunlight. To develop drugs for treating Alzheimer's disease (AD) on the basis of the amyloid cascade hypothesis, the compounds (1-4) obtained by photoreaction were evaluated for BACE1 inhibitory activity. The hydrolysates (5 and 6) of polyozellin, the main secondary metabolites of P. multiplex, were also evaluated. The photoreaction products (3 and 4) and hydrolysates (5 and 6) of polyozellin showed BACE1 inhibitory activity (IC50: 2.2, 16.4, 23.3, and 5.3 μM, respectively).
Collapse
Affiliation(s)
- Hayato Otsuka
- Department of Pharmacognosy and Phytochemistry, Meiji Pharmaceutical University, Noshio 2-522-1, Kiyose-Shi, Tokyo, 204-8588, Japan
| | - Keiyo Nakai
- Department of Chemistry, Chemical R&D Laboratory, SPERA PHARMA, Inc, 17-85, Jusohonmachi 2-Chome, Yodogawa-Ku, Osaka, 532-0024, Japan
| | - Emi Shimizu
- Department of Pharmacognosy and Phytochemistry, Meiji Pharmaceutical University, Noshio 2-522-1, Kiyose-Shi, Tokyo, 204-8588, Japan
| | - Takamasa Yamaguchi
- Department of Pharmacognosy and Phytochemistry, Meiji Pharmaceutical University, Noshio 2-522-1, Kiyose-Shi, Tokyo, 204-8588, Japan
| | - Mitsuhisa Yamano
- Department of Chemistry, Chemical R&D Laboratory, SPERA PHARMA, Inc, 17-85, Jusohonmachi 2-Chome, Yodogawa-Ku, Osaka, 532-0024, Japan
| | - Hiroaki Sasaki
- Department of Pharmacognosy and Phytochemistry, Meiji Pharmaceutical University, Noshio 2-522-1, Kiyose-Shi, Tokyo, 204-8588, Japan
| | - Kiyotaka Koyama
- Department of Pharmacognosy and Phytochemistry, Meiji Pharmaceutical University, Noshio 2-522-1, Kiyose-Shi, Tokyo, 204-8588, Japan
| | - Kaoru Kinoshita
- Department of Pharmacognosy and Phytochemistry, Meiji Pharmaceutical University, Noshio 2-522-1, Kiyose-Shi, Tokyo, 204-8588, Japan.
| |
Collapse
|
3
|
Lescano LE, Salazar MO, Furlan RLE. Chemically engineered essential oils prepared through thiocyanation under solvent-free conditions: chemical and bioactivity alteration. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:35. [PMID: 38822174 PMCID: PMC11143095 DOI: 10.1007/s13659-024-00456-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/17/2024] [Indexed: 06/02/2024]
Abstract
The generation of chemically engineered essential oils (CEEOs) prepared from bi-heteroatomic reactions using ammonium thiocyanate as a source of bioactive compounds is described. The impact of the reaction on the chemical composition of the mixtures was qualitatively demonstrated through GC-MS, utilizing univariate and multivariate analysis. The reaction transformed most of the components in the natural mixtures, thereby expanding the chemical diversity of the mixtures. Changes in inhibition properties between natural and CEEOs were demonstrated through acetylcholinesterase TLC autography, resulting in a threefold increase in the number of positive events due to the modification process. The chemically engineered Origanum vulgare L. essential oil was subjected to bioguided fractionation, leading to the discovery of four new active compounds with similar or higher potency than eserine against the enzyme. The results suggest that the directed chemical transformation of essential oils can be a valuable strategy for discovering new acetylcholinesterase (AChE) inhibitors.
Collapse
Affiliation(s)
- Liz E Lescano
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Suipacha 531, 2000, Rosario, Argentina
| | - Mario O Salazar
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Suipacha 531, 2000, Rosario, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha 531, 2000, Rosario, Argentina.
| | - Ricardo L E Furlan
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Suipacha 531, 2000, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha 531, 2000, Rosario, Argentina
| |
Collapse
|
4
|
Beato A, Haudecoeur R, Boucherle B, Peuchmaur M. Expanding Chemical Frontiers: Approaches for Generating Diverse and Bioactive Natural Product-Like Compounds Libraries from Extracts. Chemistry 2024; 30:e202304166. [PMID: 38372433 DOI: 10.1002/chem.202304166] [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: 12/14/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024]
Abstract
The realms of natural products and synthetic compounds exhibit distinct chemical spaces that not only differ but also complement each other. While the convergence of these two domains has been explored through semisynthesis and conventional pharmacomodulation endeavours applied to natural frameworks, a recent and innovative approach has emerged that involves the combinatorial generation of libraries of 'natural product-like compounds' (NPLCs) through the direct synthetic derivatization of natural extracts. This has led to the production of numerous NPLCs that incorporate structural elements from both their natural (multiple saturated rings, oxygen content, chiral centres) and synthetic (aromatic rings, nitrogen and halogen content, drug-like properties) precursors. Through careful selection of extracts and reagents, specific bioactivities have been achieved, and this strategy has been deployed in various ways, showing great promise without reaching its full potential to date. This review seeks to provide an overview of reported examples involving the chemical engineering of extracts, showcasing a spectrum of natural product alterations spanning from simple substitutions to complete scaffold remodelling. It also includes an analysis of the accomplishments, perspectives and technical challenges within this field.
Collapse
Affiliation(s)
- Aurélien Beato
- Univ. Grenoble Alpes, CNRS, DPM, Bâtiment E Pôle Chimie BP 53, 38000, Grenoble, France
| | - Romain Haudecoeur
- Univ. Grenoble Alpes, CNRS, DPM, Bâtiment E Pôle Chimie BP 53, 38000, Grenoble, France
| | - Benjamin Boucherle
- Univ. Grenoble Alpes, CNRS, DPM, Bâtiment E Pôle Chimie BP 53, 38000, Grenoble, France
| | - Marine Peuchmaur
- Univ. Grenoble Alpes, CNRS, DPM, Bâtiment E Pôle Chimie BP 53, 38000, Grenoble, France
| |
Collapse
|
5
|
Suzuki Y, Ichinohe K, Sugawara A, Kida S, Murase S, Zhang J, Yamada O, Hattori T, Oshima Y, Kikuchi H. Development of Indole Alkaloid-Type Dual Immune Checkpoint Inhibitors Against CTLA-4 and PD-L1 Based on Diversity-Enhanced Extracts. Front Chem 2021; 9:766107. [PMID: 34858943 PMCID: PMC8630621 DOI: 10.3389/fchem.2021.766107] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
Cancer immunotherapy involves the use of the immune system for cancer treatment. Recently, immune checkpoint-blocking antibodies have become integral for the treatment of some cancers. However, small molecules exhibit advantages over monoclonal antibody drugs, such as cell penetration, long half-life, and low manufacturing costs, and the possibility of oral administration. Thus, it is imperative to develop small-molecule immune checkpoint inhibitors. Previously, we have screened a library of synthetic indole-alkaloid-type compounds, which are produced by diversity-enhanced extracts of Japanese cornelian cherry, and reported that an unnatural pentacyclic compound inhibits CTLA-4 gene expression. In this study, immune checkpoint inhibitors with increased potency were developed by introducing substituents and conversion of functional groups based on the unnatural pentacyclic compound. The developed compounds suppressed not only CTLA-4 and PD-L1 gene expression but also protein expression on the cell surface. Their efficacy was not as potent as that of the existing small-molecule immune checkpoint inhibitors, but, to the best of our knowledge, the developed compounds are the first reported dual small-molecule inhibitors of CTLA-4 and PD-L1.
Collapse
Affiliation(s)
- Yoshihide Suzuki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Keisuke Ichinohe
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Akihiro Sugawara
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Shinya Kida
- Research and Development Center, FUSO Pharmaceutical Industries, Ltd., Osaka, Japan
| | - Shinya Murase
- Research and Development Center, FUSO Pharmaceutical Industries, Ltd., Osaka, Japan
| | - Jing Zhang
- Research and Development Center, FUSO Pharmaceutical Industries, Ltd., Osaka, Japan
| | - Osamu Yamada
- Research and Development Center, FUSO Pharmaceutical Industries, Ltd., Osaka, Japan
| | - Toshio Hattori
- Research Institute of Health and Welfare, Kibi International University, Takahashi, Japan
| | - Yoshiteru Oshima
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Haruhisa Kikuchi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.,Division of Natural Medicines, Faculty of Pharmacy, Keio University, Tokyo, Japan
| |
Collapse
|
6
|
Nazir M, Saleem M, Tousif MI, Anwar MA, Surup F, Ali I, Wang D, Mamadalieva NZ, Alshammari E, Ashour ML, Ashour AM, Ahmed I, Elizbit, Green IR, Hussain H. Meroterpenoids: A Comprehensive Update Insight on Structural Diversity and Biology. Biomolecules 2021; 11:957. [PMID: 34209734 PMCID: PMC8301922 DOI: 10.3390/biom11070957] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 11/17/2022] Open
Abstract
Meroterpenoids are secondary metabolites formed due to mixed biosynthetic pathways which are produced in part from a terpenoid co-substrate. These mixed biosynthetically hybrid compounds are widely produced by bacteria, algae, plants, and animals. Notably amazing chemical diversity is generated among meroterpenoids via a combination of terpenoid scaffolds with polyketides, alkaloids, phenols, and amino acids. This review deals with the isolation, chemical diversity, and biological effects of 452 new meroterpenoids reported from natural sources from January 2016 to December 2020. Most of the meroterpenoids possess antimicrobial, cytotoxic, antioxidant, anti-inflammatory, antiviral, enzyme inhibitory, and immunosupressive effects.
Collapse
Affiliation(s)
- Mamona Nazir
- Department of Chemistry, Government Sadiq College Women University Bahawalpur, Bahawalpur 63100, Pakistan
| | - Muhammad Saleem
- Institute of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Muhammad Imran Tousif
- Department of Chemistry, DG Khan Campus, University of Education Lahore, Dera Ghazi Khan 32200, Pakistan
| | - Muhammad Aijaz Anwar
- Pharmaceutical Research Division, PCSIR Laboratories Complex Karachi, Karachi 75280, Pakistan
| | - Frank Surup
- Microbial Drugs, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Iftikhar Ali
- School of Pharmaceutical Sciences and Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Department of Chemistry, Karakoram International University, Gilgit 15100, Pakistan
| | - Daijie Wang
- School of Pharmaceutical Sciences and Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Nilufar Z Mamadalieva
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D-06120 Halle, Germany
- Institute of the Chemistry of Plant Substances, Uzbekistan Academy of Sciences, Mirzo Ulugbek Str 77, Tashkent 100170, Uzbekistan
| | - Elham Alshammari
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Mohamed L Ashour
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Ahmed M Ashour
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, P.O. Box 13578, Makkah 21955, Saudi Arabia
| | - Ishtiaq Ahmed
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 1TN, UK
| | - Elizbit
- Department of Materials Engineering, National University of Sciences and Technology (NUST) H12, Islamabad 44000, Pakistan
| | - Ivan R Green
- Department of Chemistry and Polymer Science, University of Stellenbosch, Private Bag X1, Matieland, Stellenbosch 7600, South Africa
| | - Hidayat Hussain
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D-06120 Halle, Germany
| |
Collapse
|
7
|
Han J, Jiang L, Zhang L, Quinn RJ, Liu X, Feng Y. Peculiarities of meroterpenoids and their bioproduction. Appl Microbiol Biotechnol 2021; 105:3987-4003. [PMID: 33937926 DOI: 10.1007/s00253-021-11312-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 02/05/2023]
Abstract
Meroterpenoids are a class of terpenoid-containing hybrid natural products with impressive structural architectures and remarkable pharmacological activities. Remarkable advances in enzymology and synthetic biology have greatly contributed to the elucidation of the molecular basis for their biosynthesis. Here, we review structurally unique meroterpenoids catalyzed by novel enzymes and unusual enzymatic reactions over the period of last 5 years. We also discuss recent progress on the biomimetic synthesis of chrome meroterpenoids and synthetic biology-driven biomanufacturing of tropolone sesquiterpenoids, merochlorins, and plant-derived meroterpenoid cannabinoids. In particular, we focus on the novel enzymes involved in the biosynthesis of polyketide-terpenoids, nonribosomal peptide-terpenoids, terpenoid alkaloids, and meroterpenoid with unique structures. The biological activities of these meroterpenoids are also discussed. The information reviewed here might provide useful clues and lay the foundation for developing new meroterpenoid-derived drugs. KEY POINTS: • Meroterpenoids possess intriguing structural features and relevant biological activities. • Novel enzymes are involved in the biosynthesis of meroterpenoids with unique structures. • Biomimetic synthesis and synthetic biology enable the construction and manufacturing of complex meroterpenoids.
Collapse
Affiliation(s)
- Jianying Han
- Griffith Institute for Drug Discovery, Griffith University, QLD, Brisbane, 4111, Australia
| | - Lan Jiang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Lixin Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Ronald J Quinn
- Griffith Institute for Drug Discovery, Griffith University, QLD, Brisbane, 4111, Australia
| | - Xueting Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| | - Yunjiang Feng
- Griffith Institute for Drug Discovery, Griffith University, QLD, Brisbane, 4111, Australia.
| |
Collapse
|
8
|
Zhang J, Muise ES, Han S, Kutchukian PS, Costet P, Zhu Y, Kan Y, Zhou H, Shah V, Huang Y, Saigal A, Akiyama TE, Shen XL, Cai TQ, Shah K, Carballo-Jane E, Zycband E, Yi L, Tian Y, Chen Y, Imbriglio J, Smith E, Devito K, Conway J, Ma LJ, Hoek M, Sebhat IK, Peier AM, Talukdar S, McLaren DG, Previs SF, Jensen KK, Pinto S. Molecular Profiling Reveals a Common Metabolic Signature of Tissue Fibrosis. CELL REPORTS MEDICINE 2020; 1:100056. [PMID: 33205063 PMCID: PMC7659620 DOI: 10.1016/j.xcrm.2020.100056] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 10/21/2019] [Accepted: 06/23/2020] [Indexed: 12/13/2022]
Abstract
Fibrosis, or the accumulation of extracellular matrix, is a common feature of many chronic diseases. To interrogate core molecular pathways underlying fibrosis, we cross-examine human primary cells from various tissues treated with TGF-β, as well as kidney and liver fibrosis models. Transcriptome analyses reveal that genes involved in fatty acid oxidation are significantly perturbed. Furthermore, mitochondrial dysfunction and acylcarnitine accumulation are found in fibrotic tissues. Substantial downregulation of the PGC1α gene is evident in both in vitro and in vivo fibrosis models, suggesting a common node of metabolic signature for tissue fibrosis. In order to identify suppressors of fibrosis, we carry out a compound library phenotypic screen and identify AMPK and PPAR as highly enriched targets. We further show that pharmacological treatment of MK-8722 (AMPK activator) and MK-4074 (ACC inhibitor) reduce fibrosis in vivo. Altogether, our work demonstrate that metabolic defect is integral to TGF-β signaling and fibrosis.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Eric S Muise
- Department of Genetics and Pharmacogenomics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Seongah Han
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Peter S Kutchukian
- Department of Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Philippe Costet
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Yonghua Zhu
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Yanqing Kan
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Haihong Zhou
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Vinit Shah
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Yongcheng Huang
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Ashmita Saigal
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Taro E Akiyama
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Xiao-Lan Shen
- Department of Safety Assessment and Laboratory Animal Resources, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Tian-Quan Cai
- Department of Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Kashmira Shah
- Department of Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Ester Carballo-Jane
- Department of Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Emanuel Zycband
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Lan Yi
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Ye Tian
- Department of PPDM, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Ying Chen
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Jason Imbriglio
- Department of Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Elizabeth Smith
- Department of Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Kristine Devito
- Department of Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - James Conway
- Department of Genetics and Pharmacogenomics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Li-Jun Ma
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Maarten Hoek
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Iyassu K Sebhat
- Department of Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Andrea M Peier
- Department of Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Saswata Talukdar
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - David G McLaren
- Department of Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Stephen F Previs
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Kristian K Jensen
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Shirly Pinto
- Department of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA.,Kallyope Inc., 430 E 29 Street, New York, NY 10016, USA
| |
Collapse
|
9
|
Solís CM, Salazar MO, Ramallo IA, García P, Furlan RLE. A Tyrosinase Inhibitor from a Nitrogen-Enriched Chemically Engineered Extract. ACS COMBINATORIAL SCIENCE 2019; 21:622-627. [PMID: 31361945 DOI: 10.1021/acscombsci.9b00064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The enzyme tyrosinase is involved in the biosynthesis of melanin and the enzymatic browning of fruits and vegetables, and therefore, its inhibitors have potential to treat hyperpigmentary disorders or to function as food antibrowning agents. The use of hydrazine monohydrate as a reagent to prepare chemically engineered extracts can lead to semisynthetic compounds that contain the portion N-N, a fragment rarely found in natural products and present in some tyrosinase inhibitors. Here, we report the tyrosinase inhibition screening of a series of chemically engineered extracts that are diversified by reaction with hydrazine. LC-MS was used to evaluate the change in composition produced by the reaction. Bioguided fractionation of the most active chemically engineered extract, prepared from Matricaria recutita L., led to the discovery of a pyrazole that inhibits tyrosinase with an IC50 value of 28.20 ± 1.13 μM. This compound was produced by a one-pot double chemical transformation of its natural precursor, which includes an unexpected selective removal of one -OH group.
Collapse
Affiliation(s)
- Carlos M. Solís
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario-CONICET, Suipacha 531, 2000, Rosario, Argentina
| | - Mario O. Salazar
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario-CONICET, Suipacha 531, 2000, Rosario, Argentina
| | - I. Ayelen Ramallo
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario-CONICET, Suipacha 531, 2000, Rosario, Argentina
| | - Paula García
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario-CONICET, Suipacha 531, 2000, Rosario, Argentina
| | - Ricardo L. E. Furlan
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario-CONICET, Suipacha 531, 2000, Rosario, Argentina
| |
Collapse
|