Naphthoquinones and Their Derivatives: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor:

In the current era, an ever-emerging threat of multidrug-resistant (MDR) pathogens pose serious health challenges to mankind. Researchers are uninterruptedly putting their efforts to design and develop alternative, innovative strategies to tackle the antibiotic resistance displayed by varied pathogens. Among several naturally derived and chemically synthesized compounds, quinones have achieved a distinct position to defeat microbial pathogens.

  • efflux pumps
  • MDR
  • ESKAPE pathogens
  • naphthoquinones
  • plasmid curing
  • reactive oxygen species
  • topoisomerase

1. Introduction

Antibiotics represent world-class, assured molecules that have captured a gigantic share in the global market to combat ever-rising and prevalent infections. Over decades, different types of antibiotics have come into medical practice. Penicillin occupied the European and U.S. markets since its discovery in 1928 by Sir Alexander Fleming, followed by its commercial production in the 1940s [1]. Further, the world was gifted with the discovery of another antibiotic, streptomycin, by Albert Schatz, Bugie, and Waksman in 1943. This antibiotic was able to inhibit bacteria, predominantly the organisms responsible for tuberculosis [2]. After the success stories of penicillin and streptomycin, a huge number of antibiotics succeeded commercially, such that the projected rise of the global antibiotic market is up to US $67.25 billion by 2026 [3].

The challenges of resistance acquired by microbial pathogens towards existing antibiotics led to the advent of new antimicrobials. Presently, healthcare sectors are severely affected due to eternally escalating antimicrobial resistance (AMR) shown by pathogenic bacteria, parasites, viruses, and fungi. This serious threat associated with public health needs urgent attention and an immediate action plan from government policymakers, as well as private industries. It is essential to note that the challenges associated with AMR have led to a substantial cost escalation for pharmaceutical and health-care products. Patients suffering from microbial infections are ultimately the victims of long-term illness, and are therefore loaded with an additional monetary burden in the form of expensive tests and drugs [4][5]. There is also an increased morbidity and mortality rate in patients. These multidrug-resistant (MDR) pathogens are therefore referred to as “Super Bugs” [6]. MDR bacterial pathogens also comprise the ESKAPE group [7]. The abbreviation ‘ESKAPE’ has been used to designate a group having Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacterspp. In the year 2008, Rice [8] had coined the terminology ‘the ESKAPE bugs’ to denote the ability of pathogens to escape from the lethal activity of antibiotics and impose severe menace to human health. These pathogens exhibit resistance to antimicrobial drugs like carbapenems, fluoroquinolones, glycopeptides, β-lactams, β-lactam–β-lactamase inhibitor combinations, lipopeptides, macrolides, tetracyclines, and polymyxins etc. [7]. In the year 2017, the World Health Organization (WHO) published a list of priority pathogens—Priority 1 (Critical), Priority 2 (High) and Priority 3 (Medium)—exhibiting resistance to antimicrobial agents [9]. These pathogens can worsen emergency situations and therefore, need urgent surveillance so that new and more effective compounds can be brought through the pipelines. Since 2015, WHO has introduced World Antibiotic Awareness Week (WAAW) to create awareness in the public community, health workforce and among policy-formulating personnel to restrict further emergence of antibiotic resistance and its spread. Since 2020, the Tripartite Executive Committee declared WAAW dates to be 18–24 November [10].

To gain AMR, pathogens acquire plasmids (R-drug resistance) or transposons and also possess multidrug efflux pumps (EPs) to drive out the drug molecules from their system [11]. Besides, other strategies are used, like (1) inactivation, alteration, or degradation of the drug by bacterial enzymes, (2) modification of drug binding sites on the bacterial cell, (3) biofilm formation (restricting the entry of the drug), and (4) reduction in intracellular drug accumulation [12]. Challenges associated with AMR encouraged researchers to explore a variety of naturally existing and chemically synthesized compounds over the decades. Since ancient times, medicinal plants have been evidenced as a great support to tackle dreaded illnesses [13]. Recent advances in the area of phytochemicals and synthesized derivatives have been looked forward to due to their multifunctional therapeutic approaches for dealing with AMR-associated challenges [14][15][16][17]. The unique structural, biological, and functional properties of naphthoquinones (NQs), along with their derivatives, have gained enormous consideration, particularly from a medicinal chemistry perspective [18]. NQs are widely distributed as natural pigments in plants, fungi, and some animals [19]. NQ derivatives bearing hydroxyl, methyl, nitrogen, sulfur, halide, phenylamino-phenylthio, or sulphide possess exceptional biological activity. Derivatives bearing hydroxyl groups are seeking more consideration due to their broad-spectrum pharmacological properties [20]. NQs possess widespread antibacterial, antiparasitic [21][22], antifungal, [23][24], antiviral [25], and antimalarial properties [26]. In the field of cancer biology, NQs are also noticeably identified for their abilities to produce reactive oxygen species (ROS) in cancer cell lines [27][28][29]. NQs are propitious candidates over the other chemotherapeutic drugs used currently. Until today, an ample number of NQ derivatives have been analyzed for their functional potential against various pathogens. This encouraged us to present a comprehensive review of the structural diversity and multifunctional potentiality of NQs in medicinal chemistry. We also shed a light on the current understanding of the mechanistic roles of NQs in combating microbial infections. We also emphasize molecular docking—a powerful approach used in predicting the interactions of NQ molecules in biological systems.

2. Structural Diversity of Naphthoquinones Entities

Structurally, NQs constitute bicyclic structures with two carbonyl groups placed either in positions 1, 4 or 1, 2. The latter case is less frequent (Figure 1A). The chemistry and biological activities of quinones are primarily dependent on the position and chemical nature of the side groups attached (R). Groups like hydrogen, hydroxyl, methyl, nitrogen, sulfur, halide, etc. are attached to the NQ’s ring structure. Generally, the presence of a hydroxyl and/or methyl group in quinone structure is found in nature. These derivatives have been reported for a broad range of applications in pharmacology [19]. Recently, NQ derivatives isolated from plant sources including lawsone, juglone, plumbagin, shikonin and lapachol (Table 1) have fascinated researchers due to their (1) abundance, (2) structural diversity, and (3) broad-spectrum therapeutic potential [20]. It is imperative to state that 1,4-NQs derivatized at the 2nd and 3rd position with different chemical groups are recurrently reported for their biological properties (Table 1). NQs having oxygen entities at 1, 4 positions in the aromatic ring exhibit promising antimicrobial properties. Sparse literature is evident on 1,2-NQs. Realizing the significance of 1,4-NQ derivatives for biological applications, the major focus of this review remains on 1,4-NQ derivatives.

Figure 1. Diversity of naphthoquinone molecules. (A): structures of chemically synthesized naphthoquinones. (B): production, purification, characterization, and biological applications of 1,4-naphthoquinone derivatives from plant material.

Table 1. Chemical structures of plant-originated naphthoquinone derivatives.

Name and Structure of the Naphthoquinone Derivative Source of Plant Material Reference
(A) Juglone: 5-Hydroxy-1,4-naphthalenedione
Coatings 11 00434 i001
Caesalpinia sappan [30]
(B) Plumbagin: 5-Hydroxy-2-methyl-naphthalene-1,4-dione
Coatings 11 00434 i002
Plumbago zeylanica [31]
Plumbago auriculata [32]
Plumbago indica [33]
(C) Lawsone: 2-Hydroxy-1,4-naphthoquinone
Coatings 11 00434 i003
Plumbago zeylanica [34]
Lawsonia inermis [35]
(D) Shikonin: 5,8-Dihydroxy-2-[(1R)-1-hydroxy-4-methyl-3-penten-1-yl]-1,4-naphthoquinone
Coatings 11 00434 i004
Arnebia euchroma [36]
Lithospermum erythrorhizon [37]
(E) Lapachol: 2-Hydroxy-3-(3-methyl-2-butenyl)-1,4-Naphthalenedione
Coatings 11 00434 i005
Tabebuia ochracea [38][39]

This entry is adapted from the peer-reviewed paper 10.3390/coatings11040434

References

  1. Gaynes, R. The discovery of Penicillin—New insights after more than 75 years of clinical use. Emerg. Infect. Dis. 2017, 23, 849–853.
  2. Horner, W.H. Streptomycin. In Biosynthesis; Springer: Berlin/Heidelberg, Germany, 1967; pp. 373–399.
  3. Global Pharma News and Resources. Available online: (accessed on 8 September 2020).
  4. Tacconelli, E.; Carrara, E.; Savoldi, A.; Harbarth, S.; Mendelson, M.; Monnet, D.L.; Pulcini, C.; Kahlmeter, G.; Kluytmans, J.; Carmeli, Y.; et al. Discovery, research, and development of new antibiotics: The WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect. Dis. 2018, 18, 318–327.
  5. Mulani, M.S.; Kamble, E.E.; Kumkar, S.N.; Tawre, M.S.; Pardesi, K.R. Emerging strategies to combat ESKAPE pathogens in the era of antimicrobial resistance: A review. Front. Microbiol. 2019, 10, 1–24.
  6. Tanwar, J.; Das, S.; Fatima, Z.; Hameed, S. Multidrug resistance: An emerging crisis. Interdiscip. Perspect. Infect. Dis. 2014, 2014, 541340.
  7. De Oliveira, D.M.P.; Forde, B.M.; Kidd, T.J.; Harris, P.N.; Schembri, M.A.; Beatson, S.A.; Paterson, D.L.; Walker, M.J. Antimicrobial resistance in ESKAPE pathogens. Clin. Microbiol. Rev. 2020, 33, e00181-19.
  8. Rice, L.B. Federal funding for the study of antimicrobial resistance in nosocomial pathogens: No ESKAPE. J. Infect. Dis. 2008, 197, 1079–1081.
  9. World Health Organization. Available online: (accessed on 10 November 2020).
  10. World Health Organization. Available online: (accessed on 16 November 2020).
  11. Nikaido, H. Multidrug resistance in bacteria. Annu. Rev. Biochem. 2009, 78, 119–146.
  12. Santajit, S.; Indrawattana, N. Mechanisms of antimicrobial resistance in ESKAPE pathogens. BioMed Res. Int. 2016, 2016, 475067.
  13. Sharifi-Rad, J.; Salehi, B.; Stojanović-Radić, Z.Z.; Fokou, P.V.T.; Sharifi-Rad, M.; Mahady, G.B.; Sharifi-Rad, M.; Masjedi, M.-R.; Lawal, T.O.; Ayatollahi, S.A.; et al. Medicinal plants used in the treatment of tuberculosis-ethnobotanical and ethnopharmacological approaches. Biotechnol. Adv. 2020, 44, 107629.
  14. Tiwari Pandey, A.; Pandey, I.; Hachenberger, Y.; Krause, B.C.; Haidar, R.; Laux, P.; Luch, A.; Singh, M.P.; Singh, A.V. Emerging paradigm against global antimicrobial resistance via bioprospecting of mushroom into novel nanotherapeutics development. Trends Food Sci. Technol. 2020, 106, 333–344.
  15. Tiwari Pandey, A.; Pandey, I.; Zamboni, P.; Gemmati, D.; Kanase, A.; Singh, A.V.; Singh, M.P. Traditional herbal remedies with a multifunctional therapeutic approach as an implication in COVID-19 associated co-infections. Coatings 2020, 10, 761.
  16. Ansari, M.H.D.; Lavhale, S.; Kalunke, R.M.; Srivastava, P.L.; Pandit, V.; Gade, S.; Yadav, S.; Laux, P.; Luch, A.; Gemmati, D.; et al. Recent advances in plant nanobionics and nanobiosensors for toxicology applications. Curr. Nanosci. 2020, 16, 27–41.
  17. Singh, V.; Kumar, V.; Kashyap, S.; Singh, A.V.; Kishore, V.; Sitti, M.; Saxena, P.S.; Srivastava, A. Graphene oxide synergistically enhances antibiotic efficacy in vancomycin-resistant Staphylococcus aureus. ACS Appl. Bio Mater. 2019, 2, 1148–1157.
  18. Kurban, S.; Deniz, N.G.; Sayil, C.; Ozyurek, M.; Guclu, K.; Stasevych, M.; Zvarych, V.; Komarovska-Porokhnyavet, O.; Novikov, V. Synthesis, antimicrobial properties, and inhibition of catalase activity of 1,4-naphtho-and benzoquinone derivatives containing N-, S-, O-substituted. Heteroat. Chem. 2019, 2019, 1658417.
  19. Sánchez-Calvo, J.M.; Barbero, G.R.; Guerrero-Vásquez, G.; Durán, A.G.; Macías, M.; Rodríguez-Iglesias, M.A.; Molinillo, J.M.; Macías, F.A. Synthesis, antibacterial and antifungal activities of naphthoquinone derivatives: A structure–activity relationship study. Med. Chem. Res. 2016, 25, 1274–1285.
  20. Mathiyazhagan, K.; Kumaran, A.; Arjun, P. Isolation of natural naphthoquinones from Juglans regia and in vitro antioxidant and cytotoxic studies of naphthoquinones and the synthetic naphthofuran derivatives. Russ. J. Bioorganic Chem. 2018, 44, 346–353.
  21. Salas, C.O.; Faúndez, M.; Morello, A.; Diego Maya, J.A.; Tapia, R. Natural and synthetic naphthoquinones active against Trypanosoma Cruzi: An initial step towards new drugs for Chagas disease. Curr. Med. Chem. 2011, 18, 144–161.
  22. Jentzsch, J.; Koko, W.S.; Al Nasr, I.S.; Khan, T.A.; Schobert, R.; Ersfeld, K.; Biersack, B. New antiparasitic bis-naphthoquinone derivatives. Chem. Biodivers. 2020, 17, e1900597.
  23. Lall, N.; Weiganand, O.; Hussein, A.A.; Meyer, J.J.M. Antifungal activity of naphthoquinones and triterpenes isolated from the root bark of Euclea natalensis. South Afr. J. Bot. 2006, 72, 579–583.
  24. Wellington, K.W.; Nyoka, N.B.; McGaw, L.J. Investigation of the antibacterial and antifungal activity of thiolated naphthoquinones. Drug Dev. Res. 2019, 80, 386–394.
  25. Tandon, V.K.; Singh, R.V.; Yadav, D.B. Synthesis and evaluation of novel 1,4-naphthoquinone derivatives as antiviral, antifungal and anticancer agents. Bioorg. Med. Chem. Lett. 2004, 14, 2901–2904.
  26. Schuck, D.C.; Ferreira, S.B.; Cruz, L.N.; Da Rocha, D.R.; Moraes, M.S.; Nakabashi, M.; Rosenthal, P.J.; Ferreira, V.F.; Garcia, C.R. Biological evaluation of hydroxynaphthoquinones as anti-malarials. Malar. J. 2013, 12, 1–6.
  27. Klaus, V.; Hartmann, T.; Gambini, J.; Graf, P.; Stahl, W.; Hartwig, A.; Klotz, L.-O. 1,4-Naphthoquinones as inducers of oxidative damage and stress signaling in HaCaT human keratinocytes. Arch. Biochem. Biophys. 2010, 496, 93–100.
  28. Liang, W.; Cai, A.; Chen, G.; Xi, H.; Wu, X.; Cui, J.; Zhang, K.; Zhao, X.; Yu, J.; Wei, B.; et al. Shikonin induces mitochondria-mediated apoptosis and enhances chemotherapeutic sensitivity of gastric cancer through reactive oxygen species. Sci. Rep. 2016, 6, 38267.
  29. Liu, C.; Shen, G.N.; Luo, Y.H.; Piao, X.J.; Jiang, X.Y.; Meng, L.Q.; Wang, Y.; Zhang, Y.; Wang, J.R.; Wang, H.; et al. Novel 1,4-naphthoquinone derivatives induce apoptosis via ROS-mediated P38/MAPK, Akt and STAT3 signaling in human hepatoma Hep3B cells. Int. J. Biochem. Cell Biol. 2018, 96, 9–19.
  30. Lim, M.-Y.; Jeon, J.-H.; Jeong, E.-Y.; Lee, C.-H.; Lee, H.-S. Antimicrobial activity of 5-hydroxy-1,4-naphthoquinone isolated from Caesalpinia sappan toward intestinal bacteria. Food Chem. 2007, 100, 1254–1258.
  31. Periasamy, H.; Iswarya, S.; Pavithra, N.; Senthilnathan, S.; Gnanamani, A. In vitro antibacterial activity of plumbagin isolated from Plumbago zeylanica L. against methicillin-resistant Staphylococcus aureus. Lett. Appl. Microbiol. 2019, 69, 41–49.
  32. Patwardhan, R.B.; Shinde, P.S.; Chavan, K.R.; Devale, A. Reversal of plasmid encoded antibiotic resistance from nosocomial pathogens by using Plumbago auriculata root extracts. Int. J. Curr. Microbiol. Appl. Sci. 2015, 2, 187–198.
  33. Kaewbumrung, S.; Panichayupakaranant, P. Antibacterial activity of plumbagin derivative-rich Plumbago indica root extracts and chemical stability. Nat. Prod. Res. 2014, 28, 835–837.
  34. Patwardhan, R.B.; Dhakephalkar, P.K.; Chopade, B.A.; Dhavale, D.D.; Bhonde, R.R. Purification and characterization of an active principle, lawsone, responsible for the plasmid curing activity of Plumbago zeylanica root extracts. Front. Microbiol. 2018, 9, 2618.
  35. Tekin, V.; Muftuler, F.Z.B.; Guldu, O.K.; Kilcar, A.Y.; Medine, E.I.; Yavuz, M.; Unak, P.; Timur, S. Biological affinity evaluation of Lawsonia inermis origin lawsone compound and its radioiodinated form via in vitro methods. J. Radioanal. Nucl. Chem. 2015, 303, 701–708.
  36. Huang, X.Y.; Fu, H.L.; Tang, H.Q.; Yin, Z.Q.; Zhang, W.; Shu, G.; Yin, L.Z.; Zhao, L.; Yan, X.R.; Lin, J.C. Optimization extraction of shikonin using ultrasound-assisted response surface methodology and antibacterial studies. Evid. Based Complement Alternat. Med. 2020, 2020, 1–4.
  37. Lee, Y.S.; Lee, D.Y.; Kim, Y.B.; Lee, S.W.; Cha, S.W.; Park, H.W.; Kim, G.S.; Kwon, D.Y.; Lee, M.H.; Han, S.H. The mechanism underlying the antibacterial activity of shikonin against methicillin-resistant Staphylococcus aureus. Evid. Based Complement Alternat. Med. 2015, 2015, 520578.
  38. Zani, C.L.; De Oliveira, A.B.; De Oliviera, G.G. Furanonaphthoquinones from Tabebuia Ochracea. Phytochemistry 1991, 30, 2379–2381.
  39. Souza, M.A.; Johann, S.; Lima, L.A.R.d.S.; Campos, F.F.; Mendes, I.C.; Beraldo, H.; Souza-Fagundes, E.M.d.; Cisalpino, P.S.; Rosa, C.A.; Alves, T.M.d.A.; et al. The antimicrobial activity of lapachol and its thiosemicarbazone and semicarbazone derivatives. Mem. Inst. Oswaldo Cruz. 2013, 108, 342–351.
  40. Widhalm, J.R.; Rhodes, D. Biosynthesis and molecular actions of specialized 1,4-naphthoquinone natural products produced by horticultural plants. Hortic. Res. 2016, 3, 1–17.
  41. Zhou, D.-Y.; Zhu, B.-W.; Wang, X.-D.; Qin, L.; Li, D.-M.; Miao, L.; Murata, Y. Stability of polyhydroxylated 1,4-naphthoquinone pigment recovered from spines of sea urchin Strongylocentrotus nudus. Int. J. Food Sci. Technol. 2012, 47, 1479–1486.
  42. Carriço, M.d.P.S.B.; do Carmo Cardoso, M.F.; Da Silva, F.D.C.; Ferreira, V.F.; Lima, E.S.; Souza, J.V.B. Antifungal activity of synthetic naphthoquinones against dermatophytes and opportunistic fungi: Preliminary mechanism-of-action tests. Ann. Clin. Microbiol. Antimicrob. 2014, 13, 26.
  43. Adusei, E.; Adosraku, R.K.; Oppong-Kyekyeku, J.; Amengor, C.D.; Jibira, Y. Resistance modulation action, time-kill kinetics assay, and inhibition of biofilm formation effects of plumbagin from Plumbago zeylanica Linn. J. Trop. Med. 2019, 2019, 31.
  44. Vukic, M.D.; Vukovic, N.L.; Djelic, G.T.; Popovic, S.L.; Zaric, M.M.; Baskic, D.D.; Krstic, G.B.; Tesevic, V.V.; Kacaniova, M.M. Antibacterial and cytotoxic activities of naphthoquinone pigments from Onosma visianii Clem. EXCLI J. 2017, 16, 73–88.
  45. Petrosyan, M.; Shcherbakova, Y.; Sahakyan, N.; Vardanyan, Z.; Poladyan, A.; Popov, Y.; Trchounian, A. Alkanna orientalis (L.) Boiss. Plant isolated cultures and antimicrobial activity of their extracts: Phenomenon, dependence on different factors and effects on some membrane-associated properties of bacteria. Plant Cell Tissue Organ Cult. 2015, 122, 727–738.
  46. Wang, J.; Cheng, Y.; Wu, R.; Jiang, D.; Bai, B.; Tan, D.; Yan, T.; Sun, X.; Zhang, Q.; Wu, Z. Antibacterial activity of juglone against Staphylococcus aureus: From apparent to proteomic. Int. J. Mol. Sci. 2016, 17, 965.
  47. Padhye, S.; Dandawate, P.; Yusufi, M.; Ahmad, A.; Sarkar, F.H. Perspectives on medicinal properties of plumbagin and its analogs. Med. Res. Rev. 2012, 32, 1131–1158.
  48. Al-Mussawi, A.A. Isolation and identification of shikonin from Arnebia Decumbens L. and its antibacterial activity. Res. J. Appl. Sci. 2010, 6, 1452–1456.
  49. Andújar, I.; Ríos, J.L.; Giner, R.M.; Recio, M.C. Pharmacological properties of shikonin-a review of literature since 2002. Planta Med. 2013, 79, 1685–1697.
  50. Aljanaby, A.J. Antibacterial Activity of an aqueous extracts of Alkanna tinctoria roots against drug resistant aerobic pathogenic bacteria isolated from patients with burns infections. Russ. Open Med. J. 2018, 7, e0104.
  51. De Almeida, E.R. Preclinical and clinical studies of lapachol and beta-lapachone. Open Nat. Prod. J. 2009, 2, 42–47.
  52. Balachandran, C.; Al-Dhabi, N.A.; Duraipandiyan, V.; Ignacimuthu, S. Bluemomycin, a new naphthoquinone derivative from Streptomyces sp. with antimicrobial and cytotoxic properties. Biotechnol. Lett. 2021, 43, 1–4.
  53. Ravichandiran, P.; Sheet, S.; Premnath, D.; Kim, A.R.; Yoo, D.J. 1,4-Naphthoquinone analogues: Potent antibacterial agents and mode of action evaluation. Molecules 2019, 24, 1437.
  54. Tandon, V.K.; Yadav, D.B.; Singh, R.V.; Chaturvedi, A.K.; Shukla, P.K. Synthesis and biological evaluation of novel (L)-α-amino acid methyl ester, heteroalkyl, and aryl substituted 1,4-naphthoquinone derivatives as antifungal and antibacterial agents. Bioorg. Med. Chem. Lett. 2005, 15, 5324–5328.
  55. Sarkar, A.; Ghosh, S.; Shaw, R.; Patra, M.M.; Calcuttawala, F.; Mukherjee, N.; Gupta, S.K.D. Mycobacterium tuberculosis thymidylate synthase (ThyX) is a target for plumbagin, a natural product with antimycobacterial activity. PLoS ONE 2020, 15, e0228657.
  56. Meah, M.S.; Lertcanawanichakul, M.; Pedpradab, P.; Lin, W.; Zhu, K.; Li, G.; Panichayupakaranant, P. Synergistic effect on anti-methicillin-resistant Staphylococcus aureus among combinations of α-mangostin-rich extract, lawsone methyl ether and ampicillin. Lett. Appl. Microbiol. 2020, 71, 510–519.
  57. Song, R.; Yu, B.; Friedrich, D.; Li, J.; Shen, H.; Krautscheid, H.; Huang, S.D.; Kim, M.-H. Naphthoquinone-derivative as a synthetic compound to overcome the antibiotic resistance of methicillin-resistant S. aureus. Commun. Biol. 2020, 3, 1–11.
  58. Linzner, N.; Fritsch, V.N.; Busche, T.; Tung, Q.N.; Loi, V.V.; Bernhardt, J.; Kalinowski, J.; Antelmann, H. The plant-derived naphthoquinone lapachol causes an oxidative stress response in Staphylococcus aureus. Free Radic. Biol. Med. 2020, 158, 126–136.
  59. Figueredo, F.G.; Ramos, I.T.L.; Paz, J.A.; Silva, T.M.S.; Camara, C.A.; Oliveira-Tintino, C.D.d.M.; Relison Tintino, S.; de Farias, P.A.M.; Coutinho, H.D.M.; Fonteles, M.M.d.F. In silico evaluation of the antibacterial and modulatory activity of lapachol and nor-lapachol derivates. Microb. Pathog. 2020, 144, 104181.
  60. Choudhari, D.; Salunke-Gawali, S.; Chakravarty, D.; Shaikh, S.R.; Lande, D.N.; Gejji, S.P.; Rao, P.K.; Satpute, S.; Puranik, V.G.; Gonnade, R. Synthesis and biological activity of imidazole based 1,4-naphthoquinones. New J. Chem. 2020, 44, 6889–6901.
  61. Choudhari, D.; Chakravarty, D.; Lande, D.N.; Parveen, S.; Gejji, S.P.; Kodam, K.M.; Salunke-Gawali, S. Crystal structures and biological activity of homologated (N)-n-alkylammonium salts of 2-bromo-3-oxido-1,4-naphthoquinone. Struct. Chem. 2019, 30, 2257–2270.
  62. Andrade, J.C.; Morais Braga, M.F.B.; Guedes, G.M.M.; Tintino, S.R.; Freitas, M.A.; Quintans, L.J.; Menezes, I.R.A.; Coutinho, H.D.M. Menadione (vitamin K) enhances the antibiotic activity of drugs by cell membrane permeabilization mechanism. Saudi J. Biol. Sci. 2017, 24, 59–64.
  63. Moreira, C.S.; Silva, A.; Novais, J.S.; Sá Figueiredo, A.M.; Ferreira, V.F.; da Rocha, D.R.; Castro, H.C. Searching for a potential antibacterial lead structure against bacterial biofilms among new naphthoquinone compounds. J. Appl. Microbiol. 2017, 122, 651–662.
  64. Janeczko, M.; Demchuk, O.M.; Strzelecka, D.; Kubiński, K.; Masłyk, M. New family of antimicrobial agents derived from 1,4-naphthoquinone. Eur. J. Med. Chem. 2016, 124, 1019–1025.
  65. Qiu, H.Y.; Wang, P.F.; Wang, Z.Z.; Luo, Y.L.; Hu, D.Q.; Qi, J.L.; Lu, G.H.; Pang, Y.J.; Yang, R.W.; Zhu, H.L.; et al. Shikonin derivatives as inhibitors of tyrosyl-TRNA synthetase: Design, synthesis and biological evaluation. RSC Adv. 2016, 6, 83003–83010.
  66. Zmantar, T.; Miladi, H.; Kouidhi, B.; Chaabouni, Y.; Slama, R.B.; Bakhrouf, A.; Mahdouani, K.; Chaieb, K. Use of juglone as antibacterial and potential efflux pump inhibitors in Staphylococcus aureus isolated from the oral cavity. Microb. Pathog. 2016, 101, 44–49.
  67. Ravichandiran, P.; Premnath, D.; Vasanthkumar, S. Synthesis, molecular docking and antibacterial evaluation of 2-(4-(4-aminophenylsulfonyl) phenylamino)-3-(thiophen-2-ylthio) naphthalene-1,4-dione derivatives. Front. Chem. Sci. Eng. 2015, 9, 46–56.
  68. Sreelatha, T.; Kandhasamy, S.; Dinesh, R.; Shruthy, S.; Shweta, S.; Mukesh, D.; Karunagaran, D.; Balaji, R.; Mathivanan, N.; Perumal, P.T. Synthesis and sar study of novel anticancer and antimicrobial naphthoquinone amide derivatives. Bioorg. Med. Chem. Lett. 2014, 24, 3647–3651.
  69. Schlievert, P.M.; Merriman, J.A.; Salgado-Pabón, W.; Mueller, E.A.; Spaulding, A.R.; Vu, B.G.; Chuang-Smith, O.N.; Kohler, P.L.; Kirby, J.R. Menaquinone analogs inhibit growth of bacterial pathogens. Antimicrob. Agents Chemother. 2013, 57, 5432–5437.
  70. Rahmoun, N.M.; Boucherit-Otmani, Z.; Boucherit, K.; Benabdallah, M.; Villemin, D.; Choukchou-Braham, N. Antibacterial and antifungal activity of lawsone and novel naphthoquinone derivatives. Med. Mal. Infect. 2012, 42, 270–275.
  71. Mathew, R.; Kruthiventi, A.K.; Prasad, J.V.; Kumar, S.P.; Srinu, G.; Chatterji, D. Inhibition of mycobacterial growth by plumbagin derivatives. Chem. Biol. Drug Des. 2010, 76, 34–42.
  72. Tandon, V.K.; Maurya, H.K.; Mishra, N.N.; Shukla, P.K. Design, synthesis and biological evaluation of novel nitrogen and sulfur containing hetero-1,4-naphthoquinones as potent antifungal and antibacterial agents. Eur. J. Med. Chem. 2009, 44, 3130–3137.
  73. Medina, L.F.C.; Hertz, P.F.; Stefani, V.; Henriques, J.A.P.; Zanotto-Filho, A.; Brandelli, A. Aminonaphthoquinone induces oxidative stress in Staphylococcus aureus. Biochem. Cell Biol. 2006, 84, 720–727.
  74. Yap, J.K.Y.; Tan, S.Y.Y.; Tang, S.Q.; Thien, V.K.; Chan, E.W.L. Synergistic antibacterial activity between 1,4-naphthoquinone and β-lactam antibiotics against methicillin-resistant Staphylococcus aureus. Microb. Drug Resist. 2021, 27, 234–240.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations