1
|
Liu XQ, Chen YJ, Zou PS, Su JC, Pan CX, Mo DL, Su GF. Synthesis of Indole-Fused Pyrazino[1,2-a]quinazolinones by Copper(I)-Catalyzed Selective Hydroamination-Cyclization of Alkynyl-tethered Quinazolinones. Chemistry 2024; 30:e202402085. [PMID: 38926940 DOI: 10.1002/chem.202402085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 06/28/2024]
Abstract
We described a copper(I)-catalyzed atom economic and selective hydroamination-cyclization of alkynyl-tethered quinazolinones to prepare a variety of indole-fused pyrazino[1,2-a]quinazolinones in good to excellent yields ranging from 39 %-99 % under mild reaction conditions. Control experiments revealed that coordination-directed method of quinazolinone moiety with copper(I) was important for the selective hydroamination-cyclization of alkynes at the N1-atom instead of N3-atom of quinazolinone. The reaction could be easily performed at gram scales and some prepared indole-fused pyrazino[1,2-a]quinazolinones with donating groups on the indole moiety showed a distinct fluorescence emission wavelength with blue shift under the acid conditions.
Collapse
Affiliation(s)
- Xiao-Qing Liu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yu Cai Road, 541004, Guilin, China
| | - Yan-Jie Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yu Cai Road, 541004, Guilin, China
| | - Pei-Sen Zou
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yu Cai Road, 541004, Guilin, China
| | - Jun-Cheng Su
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yu Cai Road, 541004, Guilin, China
| | - Cheng-Xue Pan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yu Cai Road, 541004, Guilin, China
| | - Dong-Liang Mo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yu Cai Road, 541004, Guilin, China
| | - Gui-Fa Su
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yu Cai Road, 541004, Guilin, China
| |
Collapse
|
2
|
Guo S, Zhou H, Huang X, Peng S, Li J, Ding B, Tao Y, Huang H. New glucosidated indole-quinazoline alkaloids from mangrove endophytic fungus Aspergillus fumigatus SAl12. Nat Prod Res 2024; 38:3028-3033. [PMID: 37157883 DOI: 10.1080/14786419.2023.2209822] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/15/2023] [Accepted: 04/28/2023] [Indexed: 05/10/2023]
Abstract
Two new glucosidated indole-containing quinazoline alkaloids designated fumigatosides G (1) and H (2) were isolated from mangrove-derived fungus Aspergillus fumigatus SAl12, together with the known analogues fumigatoside B (3) and fumiquinazoline J (4). The planar structures of the new compounds were elucidated by HR-MS and NMR spectroscopic data analyses. The absolute configurations were determined by comparison of electronic circular dichroic (ECD) spectra with that of the known compound fumigatoside B and with the calculated ECD spectrum. All these indole-quinazoline compounds were tested for anti-bacterial and cytotoxic activities.
Collapse
Affiliation(s)
- Shili Guo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Haiming Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xin Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Siyan Peng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiayi Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Bo Ding
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yiwen Tao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hongbo Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Sundararajan S, Karunakaran K, Muniyan R. Structure based virtual screening and discovery of novel inhibitors against FabD protein of Mycobacterium tuberculosis. J Biomol Struct Dyn 2024; 42:6280-6291. [PMID: 37424186 DOI: 10.1080/07391102.2023.2233622] [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: 03/03/2023] [Accepted: 07/01/2023] [Indexed: 07/11/2023]
Abstract
The highly flexible nature of Mycobacterium tuberculosis (Mtb) can be owed to its tough cell wall and multiple gene interaction system which makes it resistant to frontline TB drugs. Mycolic acids are the key components of the unique cell wall that protects the organism from external threats. Proteins of the fatty acid synthesis pathway are evolutionarily conserved that enables cellular survival in harsh conditions and hence have become attractive targets. Malonyl Co-A Acyl carrier protein transacylase (FabD; MCAT, EC2.3.1.39) is an enzyme in the branching point of the unique and vast fatty acid synthase (FAS-I and FAS-II) systems of Mtb. In the present investigation, in-silico structure based drug discovery with the compounds from an open source library (NPASS) is used for target fishing and employed to understand the interaction with the target protein FabD. The potential hit compounds were filtered using exhaustive docking, considering the binding energy, key residue interaction and drug likeness property. Three compounds from the library namely NPC475074 (Hit 1), NPC260631 (Hit 2) and NPC313985 (Hit 3) with binding energies -14.45, -13.29 and -12.37 respectively were taken for molecular dynamic simulation. The results suggested that Hit 3 (NPC313985) has stable interaction with FabD protein. This article further elaborates the interaction of the identified novel compounds Hit 1 and Hit 3 along with the other known compound (Hit 2) against Mtb FabD protein. The hit compounds identified from this study could be further evaluated against mutated FabD protein and considered for in-vitro evaluation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sadhana Sundararajan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Keerthana Karunakaran
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Rajiniraja Muniyan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
4
|
Gómez-Ayuso J, Pertejo P, Hermosilla T, Carreira-Barral I, Quesada R, García-Valverde M. Harnessing unprotected deactivated amines and arylglyoxals in the Ugi reaction for the synthesis of fused complex nitrogen heterocycles. Beilstein J Org Chem 2024; 20:1758-1766. [PMID: 39076301 PMCID: PMC11285059 DOI: 10.3762/bjoc.20.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024] Open
Abstract
Piperazines and diazepines are examples of nitrogen heterocycles present in many marketed drugs highlighting their importance in the discovery of novel bioactive compounds. However, their synthesis often faces challenges, including complex functionalization and lengthy reaction sequences. Multicomponent reactions, notably the Ugi reaction, have emerged as powerful tools to address these hurdles. Here, we have demonstrated the possibility of using the combination of arylglyoxals and carboxylic acids tethered to nonprotected deactivated amines as a powerful strategy for the synthesis of complex fused heterocycles. The limited nucleophilic character of the amino group of the anthranilic acid, indole-2-carboxylic acid, pyrrole-2-carboxylic acid or N-phenylglycine has allowed the use of these compounds in the Ugi reaction without triggering competitive reactions. The additional functional group present in the resulting Ugi adduct can be leveraged in different post-condensation strategies to easily generate multiple fused nitrogen heterocycles including benzodiazepinone and piperazinone cores.
Collapse
Affiliation(s)
| | - Pablo Pertejo
- Departamento de Química, Universidad de Burgos, Burgos 09001, Spain
| | - Tomás Hermosilla
- Departamento de Química, Universidad de Burgos, Burgos 09001, Spain
| | | | - Roberto Quesada
- Departamento de Química, Universidad de Burgos, Burgos 09001, Spain
| | | |
Collapse
|
5
|
Shende VV, Bauman KD, Moore BS. The shikimate pathway: gateway to metabolic diversity. Nat Prod Rep 2024; 41:604-648. [PMID: 38170905 PMCID: PMC11043010 DOI: 10.1039/d3np00037k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Covering: 1997 to 2023The shikimate pathway is the metabolic process responsible for the biosynthesis of the aromatic amino acids phenylalanine, tyrosine, and tryptophan. Seven metabolic steps convert phosphoenolpyruvate (PEP) and erythrose 4-phosphate (E4P) into shikimate and ultimately chorismate, which serves as the branch point for dedicated aromatic amino acid biosynthesis. Bacteria, fungi, algae, and plants (yet not animals) biosynthesize chorismate and exploit its intermediates in their specialized metabolism. This review highlights the metabolic diversity derived from intermediates of the shikimate pathway along the seven steps from PEP and E4P to chorismate, as well as additional sections on compounds derived from prephenate, anthranilate and the synonymous aminoshikimate pathway. We discuss the genomic basis and biochemical support leading to shikimate-derived antibiotics, lipids, pigments, cofactors, and other metabolites across the tree of life.
Collapse
Affiliation(s)
- Vikram V Shende
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Katherine D Bauman
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Bradley S Moore
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, 92093, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
6
|
Han G, Zhang W, Acs E, Paquin A, Ronzon Q, Casaretto N, Nay B. Total Synthesis of Cyclotripeptidic Natural Products Anacine, Aurantiomide C, Polonimides A and C, and Verrucine F. Org Lett 2024; 26:2629-2634. [PMID: 38529937 DOI: 10.1021/acs.orglett.4c00658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
The total synthesis of cyclotripeptidic natural products possessing a central piperazino[2,1-b]quinazolin-3,6-dione core is described through an original strategy involving the pivotal cyclocondensation of an electrophilic homoserine lactone intermediate. The alkylidene group was spontaneously installed by autoxidation during the cyclocondensation process, while the propionamide side chain was introduced through the nickel-catalyzed aminocarbonylation of a bromoethyl intermediate. This last reaction is unprecedented on such highly functionalized intermediates. Finally, we explored structural modifications and interconversions of the natural products. Overall, this work led to anacine, aurantiomide C, polonimides A and C, and verrucine F.
Collapse
Affiliation(s)
- Guanghui Han
- Laboratoire de Synthèse Organique, Ecole Polytechnique, ENSTA Paris, CNRS, Institut Polytechnique de Paris, Route de Saclay, 91128 Palaiseau Cedex, France
| | - Wei Zhang
- Laboratoire de Synthèse Organique, Ecole Polytechnique, ENSTA Paris, CNRS, Institut Polytechnique de Paris, Route de Saclay, 91128 Palaiseau Cedex, France
| | - Emmanuelle Acs
- Laboratoire de Synthèse Organique, Ecole Polytechnique, ENSTA Paris, CNRS, Institut Polytechnique de Paris, Route de Saclay, 91128 Palaiseau Cedex, France
| | - Alexis Paquin
- Laboratoire de Synthèse Organique, Ecole Polytechnique, ENSTA Paris, CNRS, Institut Polytechnique de Paris, Route de Saclay, 91128 Palaiseau Cedex, France
| | - Quentin Ronzon
- Laboratoire de Synthèse Organique, Ecole Polytechnique, ENSTA Paris, CNRS, Institut Polytechnique de Paris, Route de Saclay, 91128 Palaiseau Cedex, France
| | - Nicolas Casaretto
- Laboratoire de Chimie Moléculaire, Ecole Polytechnique, CNRS, Institut Polytechnique de Paris, Route de Saclay, 91128 Palaiseau Cedex, France
| | - Bastien Nay
- Laboratoire de Synthèse Organique, Ecole Polytechnique, ENSTA Paris, CNRS, Institut Polytechnique de Paris, Route de Saclay, 91128 Palaiseau Cedex, France
| |
Collapse
|
7
|
Martín JF, Liras P. Targeting of Specialized Metabolites Biosynthetic Enzymes to Membranes and Vesicles by Posttranslational Palmitoylation: A Mechanism of Non-Conventional Traffic and Secretion of Fungal Metabolites. Int J Mol Sci 2024; 25:1224. [PMID: 38279221 PMCID: PMC10816013 DOI: 10.3390/ijms25021224] [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: 12/08/2023] [Revised: 12/30/2023] [Accepted: 01/09/2024] [Indexed: 01/28/2024] Open
Abstract
In nature, the formation of specialized (secondary) metabolites is associated with the late stages of fungal development. Enzymes involved in the biosynthesis of secondary metabolites in fungi are located in distinct subcellular compartments including the cytosol, peroxisomes, endosomes, endoplasmic reticulum, different types of vesicles, the plasma membrane and the cell wall space. The enzymes traffic between these subcellular compartments and the secretion through the plasma membrane are still unclear in the biosynthetic processes of most of these metabolites. Recent reports indicate that some of these enzymes initially located in the cytosol are later modified by posttranslational acylation and these modifications may target them to membrane vesicle systems. Many posttranslational modifications play key roles in the enzymatic function of different proteins in the cell. These modifications are very important in the modulation of regulatory proteins, in targeting of proteins, intracellular traffic and metabolites secretion. Particularly interesting are the protein modifications by palmitoylation, prenylation and miristoylation. Palmitoylation is a thiol group-acylation (S-acylation) of proteins by palmitic acid (C16) that is attached to the SH group of a conserved cysteine in proteins. Palmitoylation serves to target acylated proteins to the cytosolic surface of cell membranes, e.g., to the smooth endoplasmic reticulum, whereas the so-called toxisomes are formed in trichothecene biosynthesis. Palmitoylation of the initial enzymes involved in the biosynthesis of melanin serves to target them to endosomes and later to the conidia, whereas other non-palmitoylated laccases are secreted directly by the conventional secretory pathway to the cell wall space where they perform the last step(s) of melanin biosynthesis. Six other enzymes involved in the biosynthesis of endocrosin, gliotoxin and fumitremorgin believed to be cytosolic are also targeted to vesicles, although it is unclear if they are palmitoylated. Bioinformatic analysis suggests that palmitoylation may be frequent in the modification and targeting of polyketide synthetases and non-ribosomal peptide synthetases. The endosomes may integrate other small vesicles with different cargo proteins, forming multivesicular bodies that finally fuse with the plasma membrane during secretion. Another important effect of palmitoylation is that it regulates calcium metabolism by posttranslational modification of the phosphatase calcineurin. Mutants defective in the Akr1 palmitoyl transferase in several fungi are affected in calcium transport and homeostasis, thus impacting on the biosynthesis of calcium-regulated specialized metabolites. The palmitoylation of secondary metabolites biosynthetic enzymes and their temporal distribution respond to the conidiation signaling mechanism. In summary, this posttranslational modification drives the spatial traffic of the biosynthetic enzymes between the subcellular organelles and the plasma membrane. This article reviews the molecular mechanism of palmitoylation and the known fungal palmitoyl transferases. This novel information opens new ways to improve the biosynthesis of the bioactive metabolites and to increase its secretion in fungi.
Collapse
Affiliation(s)
- Juan F. Martín
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain;
| | | |
Collapse
|
8
|
Liu Y, Dietl MC, Heckershoff R, Han C, Shi H, Rudolph M, Rominger F, Caligiuri I, Asif K, Adeel M, Scattolin T, Hashmi ASK. Gold-Catalyzed Formal [4+2] Cycloaddition as Access to Antitumor-Active Spirocyclic Oxindoles from Alkynes and Isatin-Derived Ketimines. Angew Chem Int Ed Engl 2023; 62:e202304672. [PMID: 37204285 DOI: 10.1002/anie.202304672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 05/20/2023]
Abstract
Due to its excellent bioactivity profile, which is increasingly utilized in pharmaceutical and synthetic chemistry, spirooxindole is an important core scaffold. We herein describe an efficient method for the construction of highly functionalized new spirooxindolocarbamates via a gold-catalyzed cycloaddition reaction of terminal alkynes or ynamides with isatin-derived ketimines. This protocol has a good functional group compatibility, uses readily available starting materials, mild reaction conditions, low catalyst loadings and no additives. It enables the transformation of various functionalized alkyne groups into cyclic carbamates. Gram-scale synthesis was achieved and DFT calculations verify the feasibility of the mechanistic proposal. Some of the target products exhibit good to excellent antiproliferative activity on human tumor cell lines. In addition, one of the most active compounds displayed a remarkable selectivity towards tumor cells over normal ones.
Collapse
Affiliation(s)
- Yaowen Liu
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Martin C Dietl
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Robin Heckershoff
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Chunyu Han
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Hongwei Shi
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Matthias Rudolph
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Frank Rominger
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Isabella Caligiuri
- Pathology Unit, Department of Molecular Biology and Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Kanwal Asif
- Pathology Unit, Department of Molecular Biology and Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174, Venezia-Mestre, Italy
| | - Muhammad Adeel
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174, Venezia-Mestre, Italy
| | - Thomas Scattolin
- Dipartimento di Scienze Chimiche, Università degli studi di Padova, via Marzolo 1, 35131, Padova, Italy
| | - A Stephen K Hashmi
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
- Chemistry Department, Faculty of Science, King Abdulaziz University (KAU), 21589, Jeddah, Saudi Arabia
| |
Collapse
|
9
|
Almeida MC, Szemerédi N, Durães F, Long S, Resende DISP, Martins da Costa P, Pinto M, Spengler G, Sousa E. Effect of Indole-Containing Pyrazino[2,1- b]quinazoline-3,6-diones in the Virulence of Resistant Bacteria. Antibiotics (Basel) 2023; 12:antibiotics12050922. [PMID: 37237825 DOI: 10.3390/antibiotics12050922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Drug resistance is rising to alarming levels, constituting one of the major threats to global health. The overexpression of efflux pumps and the formation of biofilms constitute two of the most common resistance mechanisms, favoring the virulence of bacteria. Therefore, the research and development of effective antimicrobial agents that can also counteract resistance mechanisms are extremely important. Pyrazino[2,1-b]quinazoline-3,6-diones, from marine and terrestrial organisms and simpler synthetic analogues, were recently disclosed by us as having relevant antimicrobial properties. In this study, using a multi-step approach, it was possible to synthesize new pyrazino[2,1-b]quinazoline-3,6-diones focusing on compounds with fluorine substituents since, to the best of our knowledge, the synthesis of fluorinated fumiquinazoline derivatives had not been attempted before. The new synthesized derivatives were screened for antibacterial activity and, along with previously synthetized pyrazino[2,1-b]quinazoline-3,6-diones, were characterized for their antibiofilm and efflux-pump-inhibiting effects against representative bacterial species and relevant resistant clinical strains. Several compounds showed relevant antibacterial activity against the tested Gram-positive bacterial species with MIC values in the range of 12.5-77 μM. Furthermore, some derivatives showed promising results as antibiofilm agents in a crystal violet assay. The results of the ethidium bromide accumulation assay suggested that some compounds could potentially inhibit bacterial efflux pumps.
Collapse
Affiliation(s)
- Mariana C Almeida
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- CIIMAR--Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| | - Nikoletta Szemerédi
- Department of Medical Microbiology, Albert Szent-Gyorgyi Health Center and Albert Szent-Gyorgyi Medical School, University of Szeged, Semmelweis utca 6, 6725 Szeged, Hungary
| | - Fernando Durães
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- CIIMAR--Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| | - Solida Long
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Department of Bioengineering, Royal University of Phnom Penh, Russian Confederation Blvd, Phnom Penh 12156, Cambodia
| | - Diana I S P Resende
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- CIIMAR--Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| | - Paulo Martins da Costa
- CIIMAR--Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Madalena Pinto
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- CIIMAR--Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| | - Gabriella Spengler
- Department of Medical Microbiology, Albert Szent-Gyorgyi Health Center and Albert Szent-Gyorgyi Medical School, University of Szeged, Semmelweis utca 6, 6725 Szeged, Hungary
| | - Emília Sousa
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- CIIMAR--Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| |
Collapse
|
10
|
Sharma S, Monga Y, Gupta A, Singh S. 2-Oxindole and related heterocycles: synthetic methodologies for their natural products and related derivatives. RSC Adv 2023; 13:14249-14267. [PMID: 37179999 PMCID: PMC10173257 DOI: 10.1039/d3ra02217j] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Natural goods, medications, and pharmaceutically active substances all contain substituted oxindoles. Generally, the C-3 stereocenter of the substituents of oxindoles and their absolute arrangement have a substantial impact on the bioactivity of these substances. In this case, the desire for contemporary probe and drug-discovery programs for the synthesis of chiral compounds using desirable scaffolds with high structural diversity further drives research in this field. Also, the new synthetic techniques are generally simple to apply for the synthesis of other similar scaffolds. Herein, we review the distinct approaches for the synthesis of diverse useful oxindole scaffolds. Specifically, the research findings on the naturally existing 2-oxindole core and a variety of synthetic compounds having a 2-oxindole core are discussed. We present an overview of the construction of oxindole-based synthetic and natural products. In addition, the chemical reactivity of 2-oxindole and its related derivatives in the presence of chiral and achiral catalysts are thoroughly discussed. The data compiled herein provides broad information related to the bioactive product design, development, and applications of 2-oxindoles and the reported techniques will be helpful for the investigation of novel reactions in the future.
Collapse
Affiliation(s)
- Shivangi Sharma
- Department of Applied Chemistry, Amity School of Engineering and Technology, Amity University Madhya Pradesh Gwalior Madhya Pradesh-474 005 India
| | - Yukti Monga
- Shyamlal College, Department of Chemistry, University of Delhi Delhi-110032 India
| | - Ashu Gupta
- Shyamlal College, Department of Chemistry, University of Delhi Delhi-110032 India
| | - Shivendra Singh
- Department of Applied Chemistry, Amity School of Engineering and Technology, Amity University Madhya Pradesh Gwalior Madhya Pradesh-474 005 India
| |
Collapse
|
11
|
Martins MS, Almeida IF, Cruz MT, Sousa E. Chronic pruritus: from pathophysiology to drug design. Biochem Pharmacol 2023; 212:115568. [PMID: 37116666 DOI: 10.1016/j.bcp.2023.115568] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
Pruritus, the most common symptom in dermatology, is an innate response capable of protecting skin against irritants. Nonetheless, when it lasts more than six weeks it is assumed to be a chronic pathology having a negative impact on people's lives. Chronic pruritus (CP) can occur in common and rare skin diseases, having a high prevalence in global population. The existing therapies are unable to counteract CP or are associated with adverse effects, so the development of effective treatments is a pressing issue. The pathophysiological mechanisms underlying CP are not yet completely dissected but, based on current knowledge, involve a wide range of receptors, namely neurokinin 1 receptor (NK1R), Janus kinase (JAK), and transient receptor potential (TRP) ion channels, especially transient receptor potential vanilloid 1 (TRPV1) and transient receptor potential ankyrin 1 (TRPA1). This review will address the relevance of these molecular targets for the treatment of CP and molecules capable of modulating these receptors that have already been studied clinically or have the potential to possibly alleviate this pathology. According to scientific and clinical literature, there is an increase in the expression of these molecular targets in the lesioned skin of patients experiencing CP when compared with non-lesioned skin, highlighting their importance for the development of potential efficacious drugs through the design of antagonists/inhibitors.
Collapse
Affiliation(s)
- Márcia S Martins
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Novo Edifício do Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; Laboratory of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Isaobel F Almeida
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO-Applied Molecular Biosciences Unit, MedTech, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Maria T Cruz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Emília Sousa
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Novo Edifício do Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; Laboratory of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
12
|
Ngo-Mback MNL, Zeuko’o Menkem E, Marco HG. Antifungal Compounds from Microbial Symbionts Associated with Aquatic Animals and Cellular Targets: A Review. Pathogens 2023; 12:617. [PMID: 37111503 PMCID: PMC10142389 DOI: 10.3390/pathogens12040617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Fungal infections continue to be a serious public health problem, leading to an estimated 1.6 million deaths annually. It remains a major cause of mortality for people with a weak or affected immune system, such as those suffering from cancer under aggressive chemotherapies. On the other hand, pathogenic fungi are counted among the most destructive factors affecting crops, causing a third of all food crop losses annually and critically affecting the worldwide economy and food security. However, the limited number currently available and the cytotoxicity of the conventional antifungal drugs, which are not yet properly diversified in terms of mode of action, in addition to resistance phenomena, make the search for new antifungals imperative to improve both human health and food protection. Symbiosis has been a crucial alternative for drug discovery, through which many antimicrobials have been discovered. This review highlights some antifungal models of a defensive symbiosis of microbial symbiont natural products derived from interacting with aquatic animals as one of the best opportunities. Some recorded compounds with supposed novel cell targets such as apoptosis could lead to the development of a multitherapy involving the mutual treatment of fungal infections and other metabolic diseases involving apoptosis in their pathogenesis pathways.
Collapse
Affiliation(s)
| | | | - Heather G. Marco
- Department of Biological Sciences, University of Cape Town, Private Bag X3, Rondebosch, Cape Town 7701, South Africa
| |
Collapse
|
13
|
Cao X, Yang D, Parvathareddy J, Chu YK, Kim EJ, Fitz-Henley JN, Li X, Lukka PB, Parmar KR, Temrikar ZH, Dhole P, Adcock RS, Gabbard J, Bansal S, Lee J, Zalduondo L, Hayes E, Stabenow J, Meibohm B, Fitzpatrick EA, Bailey K, Campos RK, Julander JG, Rossi SL, Chung D, Jonsson CB, Golden JE. Efficacy of a brain-penetrant antiviral in lethal Venezuelan and eastern equine encephalitis mouse models. Sci Transl Med 2023; 15:eabl9344. [PMID: 37043558 DOI: 10.1126/scitranslmed.abl9344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 03/24/2023] [Indexed: 04/14/2023]
Abstract
Venezuelan and eastern equine encephalitis viruses (VEEV and EEEV, respectively) are mosquito-borne, neuroinvasive human pathogens for which no FDA-approved therapeutic exists. Besides the biothreat posed by these viruses when aerosolized, arthropod transmission presents serious health risks to humans, as demonstrated by the 2019 outbreak of EEE disease in the United States that resulted in 38 confirmed cases, 19 deaths, and neurological effects in survivors. Here, we describe the discovery of a 2-pyrrolidinoquinazolinone scaffold, efficiently synthesized in two to five steps, whose structural optimization resulted in profound antiviral activity. The lead quinazolinone, BDGR-49, potently reduced cellular VEEV and EEEV titers by >7 log at 1 μM and exhibited suitable intravenous and oral pharmacokinetic profiles in BALB/c mice to achieve excellent brain exposure. Outstanding in vivo efficacy was observed in several lethal, subcutaneous infection mouse models using an 8-day dosing regimen. Prophylactically administered BDGR-49 at 25 mg kg-1 per day fully protected against a 10× LD50 VEEV Trinidad donkey (TrD) challenge in BALB/c mice. Similarly, we observed 70% protection when 10× LD50 EEEV FL93-939-infected C57BL/6 mice were treated prophylactically with BDGR-49 at 50 mg kg-1 per day. Last, we observed 100% therapeutic efficacy when mice, challenged with 10× LD50 VEEV TrD, were dosed at 48 hours after infection with BDGR-49 at 25 mg kg-1 per day. Mouse brain viral titers at 96 hours after infection were reduced to values near the limit of detection. Collectively, these results underscore the substantial development potential of a well-tolerated, brain-penetrant lead compound that shows promise in preventing and treating encephalitic alphavirus disease.
Collapse
Affiliation(s)
- Xufeng Cao
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dong Yang
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jyothi Parvathareddy
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yong-Kyu Chu
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Eun Jung Kim
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Jhewelle N Fitz-Henley
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Xiaoyu Li
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Pradeep B Lukka
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Keyur R Parmar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zaid H Temrikar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Priya Dhole
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Robert Scott Adcock
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Jon Gabbard
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Shruti Bansal
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jasper Lee
- Departments of Microbiology, Immunology, Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Lillian Zalduondo
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ernestine Hayes
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jennifer Stabenow
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Elizabeth A Fitzpatrick
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Departments of Microbiology, Immunology, Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Kevin Bailey
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Rafael K Campos
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Justin G Julander
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Shannan L Rossi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Donghoon Chung
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Colleen B Jonsson
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Departments of Microbiology, Immunology, Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jennifer E Golden
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
14
|
Kim Y, Kim SY, Kim SG. Organocatalytic Asymmetric [3 + 2]-Annulations of γ-Sulfonamido/γ-Hydroxy-α,β-Unsaturated Ketones with Cyclic N-Sulfimines: Synthesis of Chiral Polyheterotricyclic Imidazolidines and Oxazolidines. J Org Chem 2023; 88:1113-1127. [PMID: 36580571 DOI: 10.1021/acs.joc.2c02634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The first organocatalytic asymmetric [3 + 2]-annulation of γ-sulfonamido-α,β-unsaturated ketones with cyclic N-sulfimines has been developed, and enantioenriched functionalized polyheterotricyclic imidazolidines were obtained in good yields and with excellent enantioselectivities. This approach was also extended to the asymmetric [3 + 2]-annulation of γ-hydroxy-α,β-unsaturated ketones, affording enantioenriched polyheterotricyclic oxazolidines. In addition, base-catalyzed [3 + 2]-annulations of γ-sulfonamido/γ-hydroxy-α,β-unsaturated ketones with cyclic N-sulfimines were re-investigated under mild reaction conditions for the synthesis of racemic polyheterotricyclic imidazolidines and oxazolidines with excellent diastereoselectivities.
Collapse
Affiliation(s)
- Yoseop Kim
- Department of Chemistry, College of Natural Science, Kyonggi University, 154-42 Gwanggyosan-ro, Yeongtong-gu, Suwon 16227, Republic of Korea
| | - Seung Yeon Kim
- Department of Chemistry, College of Natural Science, Kyonggi University, 154-42 Gwanggyosan-ro, Yeongtong-gu, Suwon 16227, Republic of Korea
| | - Sung-Gon Kim
- Department of Chemistry, College of Natural Science, Kyonggi University, 154-42 Gwanggyosan-ro, Yeongtong-gu, Suwon 16227, Republic of Korea
| |
Collapse
|
15
|
Fitz-Henley JN, Rozema SD, Golden JE. Dihydropyrazinoquinazolinones via S N2 Sulfamidate Ring-Opening and a Sequential Quinazolinone-Amidine Rearrangement Strategy (SQuAReS). J Org Chem 2022; 87:14889-14898. [PMID: 36194836 PMCID: PMC9795801 DOI: 10.1021/acs.joc.2c01717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A synthesis of dihydropyrazino-[2,1-b]-quinazolinones is described using a 2-alkylaminoquinazolinone-mediated ring opening of a-/chiral sulfamidates, followed by a tandem quinazolinone-amidine rearrangement termed SQuAReS. This approach takes advantage of sulfamidates whose regioselective ring opening, after hydrolysis, appends an optimally distanced nucleophilic amine to a quinazolinone such that subsequent domino rearrangements are favored, integrating unique substitution patterns on a privileged core. This three-step protocol integrated five telescoped transformations and generated 20 pyrazinoquinazolinones in up to 74% yield with high enantiomeric fidelity and diastereoselectivity.
Collapse
Affiliation(s)
- Jhewelle N Fitz-Henley
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin─Madison, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Soren D Rozema
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin─Madison, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Jennifer E Golden
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin─Madison, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| |
Collapse
|
16
|
Natural quinazolinones: From a treasure house to promising anticancer leads. Eur J Med Chem 2022; 245:114915. [DOI: 10.1016/j.ejmech.2022.114915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
|
17
|
Yao S, Wei C, Lin H, Zhang P, Liu Y, Deng Y, Huang Q, Xie B. Cystathionine Gamma-Lyase Regulate Psilocybin Biosynthesis in Gymnopilus dilepis Mushroom via Amino Acid Metabolism Pathways. J Fungi (Basel) 2022; 8:870. [PMID: 36012858 PMCID: PMC9410116 DOI: 10.3390/jof8080870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 12/02/2022] Open
Abstract
As a potential medicine for the treatment of depression, psilocybin has gradually attracted attention. To elucidate the molecular mechanism regulating psilocybin synthesis in Gymnopilus dilepis, ultra-performance liquid chromatography (UPLC) was used to detect the changes in psilocybin content after S-adenosyl-l-homocysteine (SAH) treatment and the changes of psilocybin content in different parts (stipe and pileus), and RNA-Seq was used to explore the mechanism of psilocybin content changes. In this study, the psilocybin content in G. dilepis mycelia treated with SAH was significantly lower than that in the control group, and the content of psilocybin in the stipe was significantly higher than that in the pileus. Transcriptome analysis revealed that differential expression genes (DEGs) were associated with cysteine and methionine metabolism. In particular, the transcription levels of genes encoding Cystathionine gamma-lyase (CTH) in different treatments and different parts were positively correlated with psilocybin content. In addition, we found that the exogenous addition of CTH activity inhibitor (DL-propargylglycine, PAG) could reduce the content of psilocybin and L-serine, and the content of psilocybin and L-serine returned to normal levels after L-cysteine supplementation, suggesting that psilocybin synthesis may be positively correlated with L-cysteine or CTH, and L-cysteine regulates the synthesis of psilocybin by affecting L-serine and 4-hydroxy-L-tryptophan. In conclusion, this study revealed a new molecular mechanism that affects psilocybin biosynthesis, which can provide a theoretical basis for improving psilocybin synthesis and the possibility for the development of biomedicine.
Collapse
Affiliation(s)
- Sen Yao
- Mycological Research Center, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chuanzheng Wei
- Mycological Research Center, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Hui Lin
- Mycological Research Center, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Peng Zhang
- Mycological Research Center, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yuanyuan Liu
- Mycological Research Center, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Youjin Deng
- Mycological Research Center, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Qianhui Huang
- College of Life Science, Ningde Normal University, Ningde 352100, China
| | - Baogui Xie
- Mycological Research Center, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
18
|
Barreiro S, Silva B, Long S, Pinto M, Remião F, Sousa E, Silva R. Fiscalin Derivatives as Potential Neuroprotective Agents. Pharmaceutics 2022; 14:pharmaceutics14071456. [PMID: 35890350 PMCID: PMC9320635 DOI: 10.3390/pharmaceutics14071456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/17/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Neurodegenerative diseases (ND) share common molecular/cellular mechanisms that contribute to their progression and pathogenesis. In this sense, we are here proposing new neuroprotection strategies by using marine-derived compounds as fiscalins. This work aims to evaluate the protective effects of fiscalin derivatives towards 1-methyl-4-phenylpyridinium (MPP+)- and iron (III)-induced cytotoxicity in differentiated SH-SY5Y cells, an in vitro disease model to study ND; and on P-glycoprotein (P-gp) transport activity, an efflux pump of drugs and neurotoxins. SH-SY5Y cells were simultaneously exposed to MPP+ or iron (III), and noncytotoxic concentrations of 18 fiscalin derivatives (0–25 μM), being the cytotoxic effect of both MPP+ and iron (III) evaluated 24 and 48 h after exposure. Fiscalins 1a and 1b showed a significant protective effect against MPP+-induced cytotoxicity and fiscalins 1b, 2b, 4 and 5 showed a protective effect against iron (III)-induced cytotoxicity. Fiscalins 4 and 5 caused a significant P-gp inhibition, while fiscalins 1c, 2a, 2b, 6 and 11 caused a modest increase in P-gp transport activity, thus suggesting a promising source of new P-gp inhibitors and activators, respectively. The obtained results highlight fiscalins with promising neuroprotective effects and with relevance for the synthesis of new derivatives for the treatment/prevention of ND.
Collapse
Affiliation(s)
- Sandra Barreiro
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (B.S.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Requimte, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Correspondence: (S.B.); (R.S.)
| | - Bárbara Silva
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (B.S.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Requimte, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Solida Long
- Department of Bioengineering, Royal University of Phnom Penh, Russian Confederation Blvd., Phnom Penh 12156, Cambodia;
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.P.); (E.S.)
| | - Madalena Pinto
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.P.); (E.S.)
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (B.S.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Requimte, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Emília Sousa
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.P.); (E.S.)
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (B.S.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Requimte, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Correspondence: (S.B.); (R.S.)
| |
Collapse
|
19
|
Chen J, Wang Y, Luo X, Chen Y. Recent research progress and outlook in agricultural chemical discovery based on quinazoline scaffold. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2022; 184:105122. [PMID: 35715060 DOI: 10.1016/j.pestbp.2022.105122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 05/08/2022] [Accepted: 05/11/2022] [Indexed: 05/27/2023]
Abstract
The discovery of new scaffolds and targets for pesticides is still a huge challenge facing the sustainable development of modern agriculture. In recent years, quinazoline derivatives have achieved great progress in drug discovery and have attracted great attention. Quinazoline is a unique bicyclic scaffold with a variety of biological activities, which increases the possibilities and flexibility of structural modification, showing enormous appeal in the discovery of new pesticides. Therefore, the agricultural biological activities, structure-activity relationships (SAR), and mechanism of action of quinazoline derivatives in the past decade were reviewed systematically, with emphasis on SAR and mechanism. Then, we prospected the application of the quinazoline scaffold as a special structure in agricultural chemical discovery, hoping to provide new ideas for the rational design and mechanism of novel quinazoline agricultural chemicals in the future.
Collapse
Affiliation(s)
- Jixiang Chen
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Huaxi District, Guiyang 550025, China.
| | - Yu Wang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Huaxi District, Guiyang 550025, China
| | - Xin Luo
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Huaxi District, Guiyang 550025, China
| | - Yifang Chen
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Huaxi District, Guiyang 550025, China
| |
Collapse
|
20
|
Skellam E. Subcellular localization of fungal specialized metabolites. Fungal Biol Biotechnol 2022; 9:11. [PMID: 35614515 PMCID: PMC9134587 DOI: 10.1186/s40694-022-00140-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/10/2022] [Indexed: 01/07/2023] Open
Abstract
Fungal specialized metabolites play an important role in the environment and have impacted human health and survival significantly. These specialized metabolites are often the end product of a series of sequential and collaborating biosynthetic enzymes that reside within different subcellular compartments. A wide variety of methods have been developed to understand fungal specialized metabolite biosynthesis in terms of the chemical conversions and the biosynthetic enzymes required, however there are far fewer studies elucidating the compartmentalization of the same enzymes. This review illustrates the biosynthesis of specialized metabolites where the localization of all, or some, of the biosynthetic enzymes have been determined and describes the methods used to identify the sub-cellular localization.
Collapse
Affiliation(s)
- Elizabeth Skellam
- Department of Chemistry and BioDiscovery Institute, University of North Texas, 1155 Union Circle, Denton, TX, 76201, USA.
| |
Collapse
|
21
|
Long S, Duarte D, Carvalho C, Oliveira R, Santarém N, Palmeira A, Resende DISP, Silva AMS, Moreira R, Kijjoa A, Cordeiro da Silva A, Nogueira F, Sousa E, Pinto MMM. Indole-Containing Pyrazino[2,1- b]quinazoline-3,6-diones Active against Plasmodium and Trypanosomatids. ACS Med Chem Lett 2022; 13:225-235. [PMID: 35178179 PMCID: PMC8842117 DOI: 10.1021/acsmedchemlett.1c00589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/07/2022] [Indexed: 01/27/2023] Open
Abstract
Malaria, leishmaniasis, and sleeping sickness are potentially fatal diseases that represent a real health risk for more than 3,5 billion people. New antiparasitic compounds are urgent leading to a constant search for novel scaffolds. Herein, pyrazino[2,1-b]quinazoline-3,6-diones containing indole alkaloids were explored for their antiparasitic potential against Plasmodium falciparum, Trypanosoma brucei, and Leishmania infantum. The synthetic libraries furnished promising hit compounds that are species specific (7, 12) or with broad antiparasitic activity (8). Structure-activity relationships were more evident for Plasmodium with anti-isomers (1S,4R) possessing excellent antimalarial activity, while the presence of a substituent on the anthranilic acid moiety had a negative effect on the activity. Hit compounds against malaria did not inhibit β-hematin, and in silico studies predicted these molecules as possible inhibitors for prolyl-tRNA synthetase both from Plasmodium and Leishmania. These results disclosed a potential new chemotype for further optimization toward novel and affordable antiparasitic drugs.
Collapse
Affiliation(s)
- Solida Long
- Laboratório
de Química Orgânica e Farmacêutica, Faculdade
de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
- Department
of Bioengineering, Royal University of Phnom
Penh, Russian Confederation
Blvd, 12156 Phnom
Penh, Cambodia
| | - Denise Duarte
- Global
Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina
Tropical, IHMT, Universidade Nova de Lisboa, 1349-008 Lisboa, Portugal
| | - Carla Carvalho
- Parasite
Disease Group, IBMC-Instituto de Biologia
Molecular e Celular, Rua Alfredo Allen, 4200-135 Porto, Portugal
| | - Rafael Oliveira
- Global
Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina
Tropical, IHMT, Universidade Nova de Lisboa, 1349-008 Lisboa, Portugal
| | - Nuno Santarém
- Parasite
Disease Group, IBMC-Instituto de Biologia
Molecular e Celular, Rua Alfredo Allen, 4200-135 Porto, Portugal
| | - Andreia Palmeira
- Laboratório
de Química Orgânica e Farmacêutica, Faculdade
de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
- CIIMAR
- Centro Interdisciplinar de Investigação Marinha e
Ambiental, Terminal de
Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| | - Diana I. S. P. Resende
- Laboratório
de Química Orgânica e Farmacêutica, Faculdade
de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
- CIIMAR
- Centro Interdisciplinar de Investigação Marinha e
Ambiental, Terminal de
Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| | - Artur M. S. Silva
- QOPNA
- Química
Orgânica, Produtos Naturais e Agroalimentares, Departamento
de Química, Universidade de Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Rui Moreira
- Research
Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculdade
de Farmácia, Universidade de Lisboa, 1649-019 Lisboa, Portugal
| | - Anake Kijjoa
- CIIMAR
- Centro Interdisciplinar de Investigação Marinha e
Ambiental, Terminal de
Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
- ICBAS-Instituto
de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Anabela Cordeiro da Silva
- Parasite
Disease Group, IBMC-Instituto de Biologia
Molecular e Celular, Rua Alfredo Allen, 4200-135 Porto, Portugal
- Departamento
de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Fátima Nogueira
- Global
Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina
Tropical, IHMT, Universidade Nova de Lisboa, 1349-008 Lisboa, Portugal
| | - Emília Sousa
- Laboratório
de Química Orgânica e Farmacêutica, Faculdade
de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
- CIIMAR
- Centro Interdisciplinar de Investigação Marinha e
Ambiental, Terminal de
Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| | - Madalena M. M. Pinto
- Laboratório
de Química Orgânica e Farmacêutica, Faculdade
de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
- CIIMAR
- Centro Interdisciplinar de Investigação Marinha e
Ambiental, Terminal de
Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| |
Collapse
|
22
|
Extremophilic Fungi from Marine Environments: Underexplored Sources of Antitumor, Anti-Infective and Other Biologically Active Agents. Mar Drugs 2022; 20:md20010062. [PMID: 35049917 PMCID: PMC8781577 DOI: 10.3390/md20010062] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 02/07/2023] Open
Abstract
Marine environments are underexplored terrains containing fungi that produce a diversity of natural products given unique environmental pressures and nutrients. While bacteria are commonly the most studied microorganism for natural products in the marine world, marine fungi are also abundant but remain an untapped source of bioactive metabolites. Given that their terrestrial counterparts have been a source of many blockbuster antitumor agents and anti-infectives, including camptothecin, the penicillins, and cyclosporin A, marine fungi also have the potential to produce new chemical scaffolds as leads to potential drugs. Fungi are more phylogenetically diverse than bacteria and have larger genomes that contain many silent biosynthetic gene clusters involved in making bioactive compounds. However, less than 5% of all known fungi have been cultivated under standard laboratory conditions. While the number of reported natural products from marine fungi is steadily increasing, their number is still significantly lower compared to those reported from their bacterial counterparts. Herein, we discuss many varied cytotoxic and anti-infective fungal metabolites isolated from extreme marine environments, including symbiotic associations as well as extreme pressures, temperatures, salinity, and light. We also discuss cultivation strategies that can be used to produce new bioactive metabolites or increase their production. This review presents a large number of reported structures though, at times, only a few of a large number of related structures are shown.
Collapse
|
23
|
Zhao H, Zhang Z, Lu W, Han P, Wang W, Jing L. 3-Carboxamide oxindoles as 1,3-C,N-bisnucleophiles for the highly diastereoselective synthesis of CF3-containing spiro-δ-lactam oxindoles featuring acyl at the ortho-position of spiro carbon atom. Tetrahedron Lett 2021. [DOI: 10.1016/j.tetlet.2021.153426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Chen PN, Hao MJ, Li HJ, Xu J, Mahmud T, Lan WJ. Biotransformations of anthranilic acid and phthalimide to potent antihyperlipidemic alkaloids by the marine-derived fungus Scedosporium apiospermum F41-1. Bioorg Chem 2021; 116:105375. [PMID: 34563999 DOI: 10.1016/j.bioorg.2021.105375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 02/07/2023]
Abstract
A new diphenylamine derivative, scediphenylamine A (1), together with six phthalimide derivatives (2-7) and ten other known compounds (8-17) were obtained from the marine-derived fungus Scedosporium apiospermum F41-1 fed with synthetically prepared anthranilic acid and phthalimide. The structure and absolute configuration of the new compound were determined by HRMS, NMR, and X-ray crystallography. Evaluation of their lipid-lowering effect in 3T3-L1 adipocytes showed that scediphenylamine A (1), N-phthaloyl-tryptophan-methyl ester (4), 5-(1,3-dioxoisoindolin-2-yl) pentanamide (5), perlolyrine (10) and flazine (11) significantly reduced triglyceride level in 3T3-L1 cells by inhibiting adipogenic differentiation and synthesis with the EC50 values of 4.39, 2.79, 3.76, 0.09, and 4.52 μM, respectively. Among them, perlolyrine (10) showed the most potent activity, making it a candidate for further development as a potential agent to treat hyperlipidemia.
Collapse
Affiliation(s)
- Pei-Nan Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Meng-Jiao Hao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Hou-Jin Li
- School of Chemistry, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Jun Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Taifo Mahmud
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Wen-Jian Lan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China.
| |
Collapse
|
25
|
New Antiproliferative Compounds against Glioma Cells from the Marine-Sourced Fungus Penicillium sp. ZZ1750. Mar Drugs 2021; 19:md19090483. [PMID: 34564145 PMCID: PMC8465473 DOI: 10.3390/md19090483] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 12/26/2022] Open
Abstract
Seven novel compounds, namely peniresorcinosides A–E (1–5), penidifarnesylin A (6), and penipyridinone A (7), together with the 11 known ones 8–17, were isolated from a culture of the marine-associated fungus Penicillium sp. ZZ1750 in rice medium. The structures of the new compounds were established based on their high-resolution electrospray ionization mass spectroscopy (HRESIMS) data, extensive nuclear magnetic resonance (NMR) spectroscopic analyses, chemical degradation, Mosher’s method, 13C-NMR calculations, electronic circular dichroism (ECD) calculations, and single crystal X-ray diffraction. Peniresorcinosides A (1) and B (2) are rare glycosylated alkylresorcinols and exhibited potent antiglioma activity, with IC50 values of 4.0 and 5.6 µM for U87MG cells and 14.1 and 9.8 µM for U251 cells, respectively.
Collapse
|
26
|
Long S, Furlani IL, de Oliveira JM, Resende DISP, Silva AMS, Gales L, Pereira JA, Kijjoa A, Cass QB, Oliveira RV, Sousa E, Pinto MMM. Determination of the Absolute Configuration of Bioactive Indole-Containing Pyrazino[2,1- b]quinazoline-3,6-diones and Study of Their In Vitro Metabolic Profile. Molecules 2021; 26:5070. [PMID: 34443658 PMCID: PMC8398919 DOI: 10.3390/molecules26165070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/02/2022] Open
Abstract
In recent decades, fungi-derived naturally occurring quinazolines have emerged as potential drug candidates. Nevertheless, most studies are conducted for bioactivity assays, and little is known about their absorption, distribution, metabolism, and elimination (ADME) properties. To perform metabolic studies, the synthesis of the naturally occurring quinazolinone, fiscalin B (1), and its chloro derivative, 4-((1H-indol-3-yl)methyl)-8,10-dichloro-1-isobutyl-1,2-dihydro-6H-pyrazino[2,1-b]quinazoline-3,6(4H)-dione (2), disclosed as an antibacterial agent, was performed in a gram scale using a microwave-assisted polycondensation reaction with 22% and 17% yields, respectively. The structure of the non-natural (+)-fiscalin B was established, for the first time, by X-ray crystallography as (1R,4S)-1, and the absolute configuration of the naturally occurring fiscalin B (-)-1 was confirmed by comparison of its calculated and experimental electronic circular dichroism (ECD) spectra as (1S,4R)-1. in vitro metabolic studies were monitored for this class of natural products for the first time by ultra-high-performance liquid chromatography (UHPLC) coupled with high-resolution mass spectrometry (HRMS). The metabolic characteristics of 1 and 2 in human liver microsomes indicated hydration and hydroxylation mass changes introduced to the parent drugs.
Collapse
Affiliation(s)
- Solida Long
- LQOF-Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (S.L.); (D.I.S.P.R.); (M.M.M.P.)
- Department of Bioegineering, Faculty of Engineering, Royal University of Phnom Penh, Russian Federation Blevd, Phnom Penh 12156, Cambodia
| | - Izadora L. Furlani
- SEPARARE–Núcleo de Pesquisa em Cromatografia, Departamento de Química, Universidade Federal de São Carlos, Rodovia Washington Luiz, km 235, São Carlos 13565-905, Brazil; (I.L.F.); (J.M.d.O.); (Q.B.C.)
| | - Juliana M. de Oliveira
- SEPARARE–Núcleo de Pesquisa em Cromatografia, Departamento de Química, Universidade Federal de São Carlos, Rodovia Washington Luiz, km 235, São Carlos 13565-905, Brazil; (I.L.F.); (J.M.d.O.); (Q.B.C.)
| | - Diana I. S. P. Resende
- LQOF-Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (S.L.); (D.I.S.P.R.); (M.M.M.P.)
- CIIMAR-Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos S/N, Matosinhos, 4450-208 Porto, Portugal;
| | - Artur M. S. Silva
- LAQV-REQUIMTE-Departamento de Química, Universidade de Aveiro, 3810-193 Aveiro, Portugal;
| | - Luís Gales
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal; (L.G.); (J.A.P.)
- i3S-IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4050-313 Porto, Portugal
| | - José A. Pereira
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal; (L.G.); (J.A.P.)
| | - Anake Kijjoa
- CIIMAR-Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos S/N, Matosinhos, 4450-208 Porto, Portugal;
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal; (L.G.); (J.A.P.)
| | - Quezia B. Cass
- SEPARARE–Núcleo de Pesquisa em Cromatografia, Departamento de Química, Universidade Federal de São Carlos, Rodovia Washington Luiz, km 235, São Carlos 13565-905, Brazil; (I.L.F.); (J.M.d.O.); (Q.B.C.)
| | - Regina V. Oliveira
- SEPARARE–Núcleo de Pesquisa em Cromatografia, Departamento de Química, Universidade Federal de São Carlos, Rodovia Washington Luiz, km 235, São Carlos 13565-905, Brazil; (I.L.F.); (J.M.d.O.); (Q.B.C.)
| | - Emília Sousa
- LQOF-Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (S.L.); (D.I.S.P.R.); (M.M.M.P.)
- CIIMAR-Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos S/N, Matosinhos, 4450-208 Porto, Portugal;
| | - Madalena M. M. Pinto
- LQOF-Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (S.L.); (D.I.S.P.R.); (M.M.M.P.)
- CIIMAR-Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos S/N, Matosinhos, 4450-208 Porto, Portugal;
| |
Collapse
|
27
|
Jaffett VA, Fitz-Henley JN, Khalifa MM, Guzei IA, Golden JE. Diastereoselective, Multicomponent Synthesis of Pyrrolopyrazinoquinazolinones via a Tandem Quinazolinone Rearrangement/Intramolecular Ring Closure of Tautomeric ( Z)-Benzamidines. Org Lett 2021; 23:5799-5803. [PMID: 34251832 PMCID: PMC8448149 DOI: 10.1021/acs.orglett.1c01955] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An expedient route to enantiopure, diastereomeric pyrrolopyrazinoquinazolinones was developed following the discovery of a domino quinazolinone rearrangement-intramolecular cyclization of N-H benzamidines. A Ugi-Mumm-Staudinger sequence employing an optically pure proline derivative gave quinazolinones that, upon N-Boc deprotection, rearranged to tautomeric Z-benzamidines. Subsequent spontaneous cyclization afforded 15 diastereomeric pyrazinoquinazolinone pairs in up to 83% overall yield and 89:11 d.r which were separated easily via routine chromatographic purification-the only one required in the entire process.
Collapse
Affiliation(s)
- Victor A. Jaffett
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, Unites States
| | - Jhewelle N. Fitz-Henley
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, Unites States
| | - Muhammad M. Khalifa
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, Unites States
| | - Ilia A. Guzei
- Molecular Structure Laboratory, Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, Unites States
| | - Jennifer E. Golden
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, Unites States
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, Unites States
| |
Collapse
|
28
|
Xu WF, Chao R, Hai Y, Guo YY, Wei MY, Wang CY, Shao CL. 17-Hydroxybrevianamide N and Its N1-Methyl Derivative, Quinazolinones from a Soft-Coral-Derived Aspergillus sp. Fungus: 13 S Enantiomers as the True Natural Products. JOURNAL OF NATURAL PRODUCTS 2021; 84:1353-1358. [PMID: 33765387 DOI: 10.1021/acs.jnatprod.1c00098] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Under the guidance of MS/MS-based molecular networking and HPLC-UV, two new alkaloid racemates, (±)-17-hydroxybrevianamide N (1) and (±)-N1-methyl-17-hydroxybrevianamide N (2), featuring a rare o-hydroxyphenylalanine residue and an imide subunit, were isolated from a soft-coral-derived Aspergillus sp. fungus. The true natural products (+)-1 and (+)-2 were further monitored and obtained from the freshly prepared EtOAc extracts, while (-)-1 and (-)-2 are artifacts generated during extraction and purification processes. Simultaneously, the structures including absolute configurations of (+)-13S-1, (-)-13R-1, (+)-13S-2, and (-)-13R-2 were elucidated on the basis of comprehensive spectroscopic analysis, ECD calculations, and X-ray diffraction data. Interestingly, basic solution promotes the racemization of (+)-1 and (-)-1, whereas acidic solution suppresses the transformation. The current research was concerned with the true natural products and their artifacts, providing critical insight into the isolation and identification of natural products.
Collapse
Affiliation(s)
- Wei-Feng Xu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Rong Chao
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Yang Hai
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Yang-Yang Guo
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Mei-Yan Wei
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Chang-Yun Wang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266200, People's Republic of China
| | - Chang-Lun Shao
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266200, People's Republic of China
| |
Collapse
|
29
|
Li X, Golden JE. Construction of N-Boc-2-Alkylaminoquinazolin-4(3 H)-Ones via a Three-Component, One-Pot Protocol Mediated by Copper(II) Chloride that Spares Enantiomeric Purity. Adv Synth Catal 2021; 363:1638-1645. [PMID: 33867902 PMCID: PMC8048503 DOI: 10.1002/adsc.202001279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/27/2020] [Indexed: 12/21/2022]
Abstract
Chiral 2-alkylquinazolinones are key synthetic intermediates, but their preparation in high optical purity is challenging. Thus, a multicomponent procedure integrating anthranilic acids, N-Boc-amino acids, and amines in the presence of methanesulfonyl chloride, N-methylimidazole, and copper(II) chloride was developed to mildly afford N-Boc-2-alkylaminoquinazolin-4(3H)-ones with excellent preservation of enantiomeric purity (>99% ee). Copper(II) chloride was essential to retaining enantiopurity, and reaction component structural changes were well tolerated, resulting in an efficient, all-in-one procedure that promotes sequential coupling, lactonization, aminolysis, and cyclization in good yields. The method was applied to the rapid assembly of four key intermediates used in the synthesis of high profile quinazolinones, including several PI3K inhibitor drugs.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Pharmaceutical SciencesSchool of PharmacyUniversity of Wisconsin-MadisonMadisonWisconsin53705-2222USA
| | - Jennifer E. Golden
- Department of Pharmaceutical SciencesSchool of PharmacyUniversity of Wisconsin-MadisonMadisonWisconsin53705-2222USA
| |
Collapse
|
30
|
Rocha MC, Fabri JHTM, Silva LP, Angolini CFF, Bertolini MC, da Cunha AF, Valiante V, Goldman GH, Fill TP, Malavazi I. Transcriptional Control of the Production of Aspergillus fumigatus Conidia-Borne Secondary Metabolite Fumiquinazoline C Important for Phagocytosis Protection. Genetics 2021; 218:6168429. [PMID: 33705521 DOI: 10.1093/genetics/iyab036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/28/2021] [Indexed: 02/04/2023] Open
Abstract
Aspergillus fumigatus produces diverse secondary metabolites whose biological functions and regulation remain to be understood. Despite the importance of the conidia for this fungus, the role of the conidia-born metabolite fumiquinazoline C (FqC) is unclear. Here, we describe a dual function of the cell-wall integrity pathway in regulating FqC biosynthesis dictated by the MAPK kinase MpkA, which phosphorylates one of the nonribosomal peptide synthetases enzymes of the cluster (FmqC), and the transcription factor RlmA, which directly regulates the expression of fmq genes. Another level of crosstalk between the FqC regulation and the cell physiology is described since the deletion of the stress-responsive transcription factor sebA provokes derepression of the fmq cluster and overproduction of FqC. Thus, we describe a mechanism by which A. fumigatus controls FqC biosynthesis orchestrated by MpkA-RlmA and SebA and hence enabling survival and adaptation to the environmental niche, given that FqC is a deterrent of ameba predation.
Collapse
Affiliation(s)
- Marina Campos Rocha
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - João Henrique Tadini Marilhano Fabri
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Lilian Pereira Silva
- Departamento de Ciências Farmacêuticas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | - Maria Célia Bertolini
- Departamento de Bioquímica e Tecnologia, Instituto de Química de Araraquara, Universidade Estadual Paulista, Araraquara, São Paulo, Brazil
| | - Anderson Ferreira da Cunha
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Vito Valiante
- Leibniz Research Group Biobricks of Microbial Natural Product Syntheses, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Gustavo Henrique Goldman
- Departamento de Ciências Farmacêuticas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Taicia Pacheco Fill
- Instituto de Química, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | | |
Collapse
|
31
|
Waghmare DS, Tambe SD, Kshirsagar UA. Pd‐Catalyzed Decarboxylative Ortho‐Aroylation of 2‐Aryl‐quinazolinone Comprising Intrinsic Directing Group with α‐Oxocarboxylic Acids. ASIAN J ORG CHEM 2020. [DOI: 10.1002/ajoc.202000487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Deepali S. Waghmare
- Department of Chemistry SP Pune University (formerly: University of Pune) Ganesh Khind Pune India
| | - Shrikant D. Tambe
- Department of Chemistry SP Pune University (formerly: University of Pune) Ganesh Khind Pune India
| | - Umesh A. Kshirsagar
- Department of Chemistry SP Pune University (formerly: University of Pune) Ganesh Khind Pune India
- Discipline of Chemistry Indian Institute of Technology Indore Simrol Indore India
| |
Collapse
|
32
|
Almeida MC, Resende DISP, da Costa PM, Pinto MMM, Sousa E. Tryptophan derived natural marine alkaloids and synthetic derivatives as promising antimicrobial agents. Eur J Med Chem 2020; 209:112945. [PMID: 33153766 DOI: 10.1016/j.ejmech.2020.112945] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/01/2020] [Accepted: 10/13/2020] [Indexed: 02/03/2023]
Abstract
Antimicrobial resistance has become a major threat to public health worldwide, as pathogenic microorganisms are finding ways to evade all known antimicrobials. Therefore, the demand for new and effective antimicrobial agents is also increasing. Natural products have always played an important role in drug discovery, either by themselves or as inspiration for synthetic compounds. The marine environment is a rich source of bioactive metabolites, and among them, tryptophan-derived alkaloids stand out for their abundance and by displaying a variety of biological activities, with antimicrobial properties being among the most significant. This review aims to reveal the potential of marine alkaloids derived from tryptophan as antimicrobial agents. Relevant examples of these compounds and their synthetic analogues reported in the last decades are presented and discussed in detail, with their mechanism of action and synthetic approaches whenever relevant. Several tryptophan-derived marine alkaloids have shown potent and promising antimicrobial activities, whether against bacteria, fungi, or virus. Synthetic approaches to many of the compounds have been developed and recent methodologies are proving to be efficient. Even though most of the studies regarding the antimicrobial activity are still preliminary, this class of compounds has proven to be worth of further investigation and may provide useful lead compounds for the development of antimicrobial agents. Overall, marine alkaloids derived from tryptophan are revealed as a valuable class of antimicrobials and molecular modifications in order to reduce the toxicity of these compounds and additional studies regarding their mechanism of action are interesting topics to explore in the future.
Collapse
Affiliation(s)
- Mariana C Almeida
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal; CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208, Matosinhos, Portugal
| | - Diana I S P Resende
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal; CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208, Matosinhos, Portugal.
| | - Paulo M da Costa
- CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208, Matosinhos, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Madalena M M Pinto
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal; CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208, Matosinhos, Portugal
| | - Emília Sousa
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal; CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208, Matosinhos, Portugal
| |
Collapse
|
33
|
Long S, Resende DISP, Palmeira A, Kijjoa A, Silva AMS, Tiritan ME, Pereira-Terra P, Freitas-Silva J, Barreiro S, Silva R, Remião F, Pinto E, Martins da Costa P, Sousa E, Pinto MMM. New marine-derived indolymethyl pyrazinoquinazoline alkaloids with promising antimicrobial profiles. RSC Adv 2020; 10:31187-31204. [PMID: 35520644 PMCID: PMC9056383 DOI: 10.1039/d0ra05319h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/11/2020] [Indexed: 01/03/2023] Open
Abstract
Due to the emergence of multidrug-resistant pathogenic microorganisms, the search for novel antimicrobials is urgent. Inspired by marine alkaloids, a series of indolomethyl pyrazino [1,2-b]quinazoline-3,6-diones was prepared using a one-pot microwave-assisted multicomponent polycondensation of amino acids. The compounds were evaluated for their antimicrobial activity against a panel of nine bacterial strains and five fungal strains. Compounds 26 and 27 were the most effective against Staphylococcus aureus ATCC 29213 reference strain with MIC values of 4 μg mL−1, and a methicillin-resistant Staphylococcus aureus (MRSA) isolate with MIC values of 8 μg mL−1. It was possible to infer that enantiomer (−)-26 was responsible for the antibacterial activity (MIC 4 μg mL−1) while (+)-26 had no activity. Furthermore, compound (−)-26 was able to impair S. aureus biofilm production and no significant cytotoxicity towards differentiated and non-differentiated SH-SY5Y cells was observed. Compounds 26, 28, and 29 showed a weak antifungal activity against Trichophyton rubrum clinical isolate with MIC 128 μg mL−1 and presented a synergistic effect with fluconazole. Indolomethyl pyrazino [1,2-b]quinazoline-3,6-diones were prepared using a one-pot multicomponent polycondensation of amino acids and were evaluated for their antimicrobial activity against a panel of nine bacterial strains and five fungal strains.![]()
Collapse
Affiliation(s)
- Solida Long
- LQOF - Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal
| | - Diana I S P Resende
- LQOF - Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal .,CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões Av. General Norton de Matos S/N 4450-208 Matosinhos Portugal
| | - Andreia Palmeira
- LQOF - Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal .,CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões Av. General Norton de Matos S/N 4450-208 Matosinhos Portugal
| | - Anake Kijjoa
- CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões Av. General Norton de Matos S/N 4450-208 Matosinhos Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal
| | - Artur M S Silva
- QOPNA - Química Orgânica, Produtos Naturais e Agroalimentares, Departamento de Química, Universidade de Aveiro 3810-193 Aveiro Portugal
| | - Maria Elizabeth Tiritan
- LQOF - Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal .,CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões Av. General Norton de Matos S/N 4450-208 Matosinhos Portugal.,CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde (IINFACTS) Rua Central de Gandra, 1317 4585-116 Gandra PRD Portugal
| | - Patrícia Pereira-Terra
- CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões Av. General Norton de Matos S/N 4450-208 Matosinhos Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal
| | - Joana Freitas-Silva
- CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões Av. General Norton de Matos S/N 4450-208 Matosinhos Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal
| | - Sandra Barreiro
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal
| | - Renata Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal
| | - Fernando Remião
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal
| | - Eugénia Pinto
- CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões Av. General Norton de Matos S/N 4450-208 Matosinhos Portugal.,Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal
| | - Paulo Martins da Costa
- CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões Av. General Norton de Matos S/N 4450-208 Matosinhos Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal
| | - Emília Sousa
- LQOF - Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal .,CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões Av. General Norton de Matos S/N 4450-208 Matosinhos Portugal
| | - Madalena M M Pinto
- LQOF - Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto Portugal .,CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões Av. General Norton de Matos S/N 4450-208 Matosinhos Portugal
| |
Collapse
|
34
|
Willems T, De Mol ML, De Bruycker A, De Maeseneire SL, Soetaert WK. Alkaloids from Marine Fungi: Promising Antimicrobials. Antibiotics (Basel) 2020; 9:antibiotics9060340. [PMID: 32570899 PMCID: PMC7345139 DOI: 10.3390/antibiotics9060340] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 01/20/2023] Open
Abstract
Resistance of pathogenic microorganisms against antimicrobials is a major threat to contemporary human society. It necessitates a perpetual influx of novel antimicrobial compounds. More specifically, Gram− pathogens emerged as the most exigent danger. In our continuing quest to search for novel antimicrobial molecules, alkaloids from marine fungi show great promise. However, current reports of such newly discovered alkaloids are often limited to cytotoxicity studies and, moreover, neglect to discuss the enigma of their biosynthesis. Yet, the latter is often a prerequisite to make them available through sufficiently efficient processes. This review aims to summarize novel alkaloids with promising antimicrobial properties discovered in the past five years and produced by marine fungi. Several discovery strategies are summarized, and knowledge gaps in biochemical production routes are identified. Finally, links between the structure of the newly discovered molecules and their activity are proposed. Since 2015, a total of 35 new antimicrobial alkaloids from marine fungi were identified, of which 22 showed an antibacterial activity against Gram− microorganisms. Eight of them can be classified as narrow-spectrum Gram− antibiotics. Despite this promising ratio of novel alkaloids active against Gram− microorganisms, the number of newly discovered antimicrobial alkaloids is low, due to the narrow spectrum of discovery protocols that are used and the fact that antimicrobial properties of newly discovered alkaloids are barely characterized. Alternatives are proposed in this review. In conclusion, this review summarizes novel findings on antimicrobial alkaloids from marine fungi, shows their potential as promising therapeutic candidates, and hints on how to further improve this potential.
Collapse
|
35
|
Review of Oxepine-Pyrimidinone-Ketopiperazine Type Nonribosomal Peptides. Metabolites 2020; 10:metabo10060246. [PMID: 32549308 PMCID: PMC7344746 DOI: 10.3390/metabo10060246] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/31/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022] Open
Abstract
Recently, a rare class of nonribosomal peptides (NRPs) bearing a unique Oxepine-Pyrimidinone-Ketopiperazine (OPK) scaffold has been exclusively isolated from fungal sources. Based on the number of rings and conjugation systems on the backbone, it can be further categorized into three types A, B, and C. These compounds have been applied to various bioassays, and some have exhibited promising bioactivities like antifungal activity against phytopathogenic fungi and transcriptional activation on liver X receptor α. This review summarizes all the research related to natural OPK NRPs, including their biological sources, chemical structures, bioassays, as well as proposed biosynthetic mechanisms from 1988 to March 2020. The taxonomy of the fungal sources and chirality-related issues of these products are also discussed.
Collapse
|
36
|
Wang Q, Chen W, Wang Q, Tao Y, Yu R, Pan G, Dang J. Preparative separation of isoquinoline alkaloids from
Corydalis impatiens
using middle chromatogram isolated gel column coupled with positively charged reversed‐phase liquid chromatography. J Sep Sci 2020; 43:2521-2528. [DOI: 10.1002/jssc.201901164] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/03/2020] [Accepted: 03/28/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Qi Wang
- Key Laboratory of Tibet Plateau Phytochemistry of Qinghai ProvinceQinghai Nationalities University Xining Qinghai P. R. China
| | - Wenjie Chen
- Key Laboratory of Tibetan Medicine ResearchNorthwest Institute of Plateau BiologyChinese Academy of Sciences Xining Qinghai P. R. China
| | - Qilan Wang
- Key Laboratory of Tibetan Medicine ResearchNorthwest Institute of Plateau BiologyChinese Academy of Sciences Xining Qinghai P. R. China
| | - Yanduo Tao
- Key Laboratory of Tibetan Medicine ResearchNorthwest Institute of Plateau BiologyChinese Academy of Sciences Xining Qinghai P. R. China
| | - Ruitao Yu
- Key Laboratory of Tibetan Medicine ResearchNorthwest Institute of Plateau BiologyChinese Academy of Sciences Xining Qinghai P. R. China
| | - Guoqing Pan
- Key Laboratory of Tibet Plateau Phytochemistry of Qinghai ProvinceQinghai Nationalities University Xining Qinghai P. R. China
| | - Jun Dang
- Key Laboratory of Tibetan Medicine ResearchNorthwest Institute of Plateau BiologyChinese Academy of Sciences Xining Qinghai P. R. China
| |
Collapse
|
37
|
Barbosa F, Pinto E, Kijjoa A, Pinto M, Sousa E. Targeting antimicrobial drug resistance with marine natural products. Int J Antimicrob Agents 2020; 56:106005. [PMID: 32387480 DOI: 10.1016/j.ijantimicag.2020.106005] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 01/10/2023]
Abstract
The rise and spread of antimicrobial resistance represents one of the most pressing health issues of today. Antimicrobial resistance in micro-organisms can arise due to a multiplicity of factors, including permeability changes in the cell membrane, increase of drug efflux pumps, enzymatic modification or inactivation of the antibiotic, target site modification, alternative metabolic pathways and biofilm formation. The marine environment is a valuable source of diverse natural products with a huge variety of biological activities. Among them, antimicrobial compounds show promising biological activities against numerous drug-resistant bacteria and fungi, making marine natural products a very promising resource in the search for novel antimicrobial agents. This review summarises the state-of-art of marine natural products with antibacterial and antifungal properties against drug-resistant micro-organisms. These natural products were categorised based on their chemical structure, and their respective sources and activities are highlighted. The chemical diversity associated with these marine-derived molecules is enormous, including peptides, polyketides, alkaloids, sterols, terpenoids, lactones, halogenated compounds, nucleosides, etc., some of which have rare substructures. Some of the marine compounds mentioned do not have intrinsic antimicrobial activity but potentiate the antimicrobial effect of other antimicrobials via inhibition of efflux pumps. Although these agents are still in preclinical studies, evidence of their in vivo efficacy suggest research of new drugs from the ocean to overcome antimicrobial resistance in order to fulfil an unmet medical need.
Collapse
Affiliation(s)
- Filipa Barbosa
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Eugénia Pinto
- Laboratory of Microbiology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal
| | - Anake Kijjoa
- CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Madalena Pinto
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal.
| |
Collapse
|
38
|
Li CJ, Chen PN, Li HJ, Mahmud T, Wu DL, Xu J, Lan WJ. Potential Antidiabetic Fumiquinazoline Alkaloids from the Marine-Derived Fungus Scedosporium apiospermum F41-1. JOURNAL OF NATURAL PRODUCTS 2020; 83:1082-1091. [PMID: 32130008 DOI: 10.1021/acs.jnatprod.9b01096] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Fumiquinazoline alkaloids have attracted much attention from medicinal and natural product chemists due to their interesting structures and biological potential. In this study, three new and 12 known fumiquinazoline alkaloids were isolated and characterized from the marine fungus Scedosporium apiospermum F41-1. The structures of the new compounds and their absolute configurations were determined using NMR spectroscopy, ECD, and OR calculations. The compounds were evaluated for their antidiabetic potential by determining their triglyceride-promoting activity using 3T3-L1 adipocytes. One of the new compounds, scequinadoline J (14), as well as scequinadolines D (9) and E (10), was found to promote triglyceride accumulation in 3T3-L1 cells. Scequinadoline D (9) demonstrated the most potent activity, with an EC50 value of 0.27 ± 0.03 μM. Quantitative polymerase chain reaction experiments suggested that scequinadoline D (9) acts through activation of the PPARγ pathway. It stimulated the mRNA expression of PPARγ, AMPKα, C/EBPα, LXRα, SCD-1, and FABP4. In addition, its triglyceride-promoting efficacy could be blocked by a double dose of the PPARγ antagonist GW9662. These results indicated that scequinadoline D (9) is a potent insulin sensitizer that targets adipocytes and may be useful for the treatment of type 2 diabetes mellitus after further investigation.
Collapse
Affiliation(s)
- Chan-Juan Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Pei-Nan Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Hou-Jin Li
- School of Chemistry, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Taifo Mahmud
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon 97331, United States
| | - Dong-Lan Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Jun Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Wen-Jian Lan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| |
Collapse
|
39
|
Guo YW, Gong BQ, Yuan J, Li HJ, Mahmud T, Huang Y, Li JF, Yang DP, Lan WJ. l-Phenylalanine Alters the Privileged Secondary Metabolite Production in the Marine-Derived Fungus Trichoderma erinaceum F1-1. JOURNAL OF NATURAL PRODUCTS 2020; 83:79-87. [PMID: 31886665 DOI: 10.1021/acs.jnatprod.9b00710] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The effects of a single-amino-acid culture strategy on secondary metabolite production in the marine-derived fungus Trichoderma erinaceum F1-1 were investigated by culturing the fungus in GPY medium supplemented or not supplemented with l-phenylalanine. A suite of secondary metabolites, including seven terpenoids (1-7) and one polyketide (8), among which are four new compounds, harziandione A (1), cyclonerodiols A and B (3, 4), and trichodermaerin A (6), were isolated from the GPY medium without l-phenylanine, whereas 18 aromatic compounds (9-26), including six new compounds, trichoderolides A-F (9, 10, and 14-17), were isolated from the culture grown in the GPY medium with l-phenylalanine. The structures of the new compounds were determined by high-resolution mass spectrometry, NMR spectroscopic analysis, optical rotation calculations, chemical methods, and X-ray crystallography. Compounds 10, 12, 13, and 26 exhibited cytotoxic activities against MDA-MB-435 human melanocyte cancer cells. Compound 26 was cytotoxic to A549 adenocarcinomic human alveolar basal epithelial cells.
Collapse
Affiliation(s)
- Yong-Wei Guo
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , People's Republic of China
| | - Ben-Qiang Gong
- School of Life Sciences , Sun Yat-sen University , Guangzhou 510006 , People's Republic of China
| | - Jie Yuan
- Zhongshan School of Medicine , Sun Yat-sen University , Guangzhou 510080 , People's Republic of China
| | - Hou-Jin Li
- School of Chemistry , Sun Yat-sen University , Guangzhou 510275 , People's Republic of China
| | - Taifo Mahmud
- Department of Pharmaceutical Sciences , Oregon State University , Corvallis , Oregon 97331 , United States
| | - Yun Huang
- School of Basic Medical Sciences , Southern Medical University , Guangzhou 510515 , People's Republic of China
| | - Jian-Feng Li
- School of Life Sciences , Sun Yat-sen University , Guangzhou 510006 , People's Republic of China
| | - De-Po Yang
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , People's Republic of China
| | - Wen-Jian Lan
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , People's Republic of China
| |
Collapse
|
40
|
Zhao Y, Liu F, He G, Li K, Zhu C, Yu W, Zhang C, Xie M, Lin J, Zhang J, Jin Y. Discovery of arylamide-5-anilinoquinazoline-8-nitro derivatives as VEGFR-2 kinase inhibitors: Synthesis, in vitro biological evaluation and molecular docking. Bioorg Med Chem Lett 2019; 29:126711. [PMID: 31668972 DOI: 10.1016/j.bmcl.2019.126711] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/23/2019] [Accepted: 09/19/2019] [Indexed: 01/07/2023]
Abstract
Herein, we embarked on a structural optimization campaign aiming at the discovery of novel anticancer agents with our previously reported XL-6f as a lead compound. A library of 23 compounds has been synthesized based on the highly conserved active site of VEGFR-2. Several title compounds exhibited selective inhibitory activities against VEGFR-2, which also displayed selective anti-proliferation potency against HepG2 cell. All synthesized compounds were evaluated for anti-angiogenesis capability. Compound 7o showed the most potent anti-angiogenesis ability, the efficient cytotoxic activities (in vitro against HUVEC and HepG2 cell lines with IC50 values of 0.58 and 0.23 µM, respectively). The molecular docking analysis revealed 7o is a Type-II inhibitor of VEGFR-2 kinase. In general, these results indicated these arylamide-5-anilinoquinazoline-8-nitro derivatives are promising inhibitors of VEGFR-2 for the potential treatment of anti-angiogenesis.
Collapse
Affiliation(s)
- Yongqiang Zhao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Feifei Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Guojing He
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Ke Li
- Biomedical Department, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, PR China.
| | - Changcheng Zhu
- Institute of Drug Research and Development, Kunming Pharmaceutical Corporation, Kunming 650100, PR China
| | - Wei Yu
- Pharmaceutical Department, Kunming General Hospital of Chengdu Military Command, Kunming 650118, PR China
| | - Conghai Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Mingjin Xie
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Jun Lin
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China.
| | - Jihong Zhang
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, PR China.
| | - Yi Jin
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China.
| |
Collapse
|
41
|
He C, Li Z, Xu J, Ren H. Asymmetric Synthesis of Spirocyclic Oxindole δ-Lactams via NHC-Catalyzed Formal [2+4] Annulation of Aliphatic Aldehydes with Oxindole-Derived α,β-Unsaturated Ketimines. J Org Chem 2019; 84:12177-12186. [PMID: 31436995 DOI: 10.1021/acs.joc.9b01760] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
An N-heterocyclic carbene (NHC)-catalyzed formal [2+4] annulation reaction of aliphatic aldehydes with oxindole-derived α,β-unsaturated ketimines under oxidative conditions is reported, affording spirocyclic oxindole δ-lactams with good yields, moderate diastereoselectivies, and good to excellent enantioselectivies. This reaction can be readily carried out on a gram scale, and the products could be further transformed to other synthetically useful compounds.
Collapse
Affiliation(s)
- Chonglong He
- Department of Chemistry , Zhejiang Sci-Tech University , Hangzhou 310018 , People's Republic of China
| | - Zhanhuan Li
- Department of Chemistry , Zhejiang Sci-Tech University , Hangzhou 310018 , People's Republic of China
| | - Jianfeng Xu
- Department of Chemistry , Zhejiang Sci-Tech University , Hangzhou 310018 , People's Republic of China
| | - Hongjun Ren
- Advanced Research Institute and Department of Chemistry , Taizhou University , 1139 Shifu Avenue , Taizhou 318000 , People's Republic of China
| |
Collapse
|
42
|
Mai P, Coby L, Li SM. Different behaviors of cyclic dipeptide prenyltransferases toward the tripeptide derivative ardeemin fumiquinazoline and its enantiomer. Appl Microbiol Biotechnol 2019; 103:3773-3781. [PMID: 30863875 DOI: 10.1007/s00253-019-09723-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/07/2019] [Accepted: 02/24/2019] [Indexed: 11/29/2022]
Abstract
In nature, cyclic dipeptide prenyltransferases catalyze regioselective Friedel-Crafts alkylations of tryptophan-containing cyclic dipeptides. This enzyme class, belonging to the dimethylallyl tryptophan synthase superfamily, is known to be flexible toward aromatic prenyl acceptors, while mostly retaining its typical regioselectivity. Ardeemin fumiquinazoline (FQ) (1), a tryptophan-containing cyclic tripeptide derivative, is assembled in Aspergillus fischeri by the non-ribosomal peptide synthetase ArdA and modified by the prenyltransferase ArdB, leading to the pharmaceutically active hexacyclic ardeemin. Therefore, 1 and its enantiomer ent-ardeemin FQ (2) constitute potential substrates for aromatic prenyltransferases. In this study, we investigated the acceptance of both enantiomers by two cyclic dipeptide C2-prenyltransferases BrePT and FtmPT1 and three C3-prenyltransferases CdpNPT, CdpC3PT, and AnaPT. LC-MS analysis of the incubation mixtures and NMR analysis of the isolated products revealed that the stereochemistry at C11 and C14 in 1 and 2 has a strong influence on their acceptance by these enzymes and the regioselectivity of the prenylation reactions. 1 was very well accepted by BrePT, FtmPT1, and CdpNPT, with C2- or C3-prenylated derivatives as predominant products, which fills the prenylation gaps by tryptophan prenyltransferases reported in a previous study. 2 was a poor substrate for all the enzymes and converted with low regioselectivity and mainly prenylated at C6 and C7 of the indole moiety.
Collapse
Affiliation(s)
| | | | - Shu-Ming Li
- Institut für Pharmazeutische Biologie und Biotechnologie, Philipps-Universität Marburg, Robert-Koch-Str. 4, 35037, Marburg, Germany.
| |
Collapse
|
43
|
Yan D, Chen Q, Gao J, Bai J, Liu B, Zhang Y, Zhang L, Zhang C, Zou Y, Hu Y. Complexity and Diversity Generation in the Biosynthesis of Fumiquinazoline-Related Peptidyl Alkaloids. Org Lett 2019; 21:1475-1479. [DOI: 10.1021/acs.orglett.9b00260] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Daojiang Yan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Qibin Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Jie Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Jian Bai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Bingyu Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Yalong Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Le Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Chen Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| | - Yi Zou
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Youcai Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, P.R. China
| |
Collapse
|
44
|
Synthesis of New Proteomimetic Quinazolinone Alkaloids and Evaluation of Their Neuroprotective and Antitumor Effects. Molecules 2019; 24:molecules24030534. [PMID: 30717179 PMCID: PMC6384550 DOI: 10.3390/molecules24030534] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 11/27/2022] Open
Abstract
New quinazolinone derivatives of the marine-derived alkaloids fiscalin B (3) and fumiquinazoline G (1), with neuroprotective and antitumor effects, were synthesized. Eleven quinazolinone-containing indole alkaloids were synthesized, proceeding the anti analogs via a one-pot method, and the syn analogs by the Mazurkiewicz-Ganesan approach. The neuroprotection capacity of these compounds on the rotenone-damage human neuroblastoma cell SH-SY5y was evaluated using the MTT assay. Compounds 1, 3, 5, and 7 showed more than 25% protection. The antitumor activity was investigated using the sulforhodamine B assay and some compounds were tested on the non-malignant MCF-12A cells. Fumiquinazoline G (1) was the most potent compound, with GI50 values lower than 20 µM. Compounds 5, 7, and 11 were more active in all tumor cell lines when compared to their enantiomers. Compounds 5, 7, 10, and 11 had very little effect in the viability of the non-malignant cells. Differences between enantiomeric pairs were also noted as being essential for these activities the S-configuration at C-4. These results reinforce the previously described activities of the fiscalin B (3) as substance P inhibitor and fumiquinazoline G (1) as antitumor agent showing potential as lead compounds for the development of drugs for treatment of neurodegenerative disorders and cancer, respectively.
Collapse
|