Antimicrobial Properties of Capsaicin: Available Data and Future Research Perspectives
Abstract
:1. Introduction
2. Antibacterial Properties of Capsaicin
2.1. Antibacterial Activity against Staphylococcus aureus
2.2. Antibacterial Activity against Group A Hemolytic Streptococci
2.3. Antimicrobial Activity against Enterococcus Species
2.4. Antimicrobial Activity against Bacillus Species
2.5. Antimicrobial Activity against Listeria monocytogenes
2.6. Antimicrobial Activity against Vibrio cholerae
2.7. Antimicrobial Activity against Acinetobacter baumanii
2.8. Antimicrobial Activity against Helicobacter pylori
2.9. Antimicrobial Activity against Salmonella typhimurium
2.10. Antibacterial Activity against Escherichia coli
2.11. Antibacterial Activity against Klebsiella pneumoniae
2.12. Antimicrobial Activity against Proteus Species
2.13. Antimicrobial Activity against Pseudomonas Species
3. Capsaicin as an Antifungal Agent
3.1. Antifungal Activity against Candida spp.
3.2. Antifungal Activity against Aspergillus parasiticus
4. Capsaicin as an Antiparasitic Agent
4.1. Antiparasitic Activity against Toxoplasma gondii
4.2. Antiparasitic Activity against Trypanosoma cruzi
5. Capsaicin as an Antiviral Agent
5.1. Antiviral Activity against the Influenza Virus
5.2. Antiviral Activity against the Lassa Virus
6. Discussion
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Antimicrobial Resistance. Available online: https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance (accessed on 26 August 2023).
- Brinkac, L.; Voorhies, A.; Gomez, A.; Nelson, K.E. The Threat of Antimicrobial Resistance on the Human Microbiome. Microb. Ecol. 2017, 74, 1001–1008. [Google Scholar] [CrossRef]
- Ojala, T.; Remes, S.; Haansuu, P.; Vuorela, H.; Hiltunen, R.; Haahtela, K.; Vuorela, P. Antimicrobial activity of some coumarin containing herbal plants growing in Finland. J. Ethnopharmacol. 2000, 73, 299–305. [Google Scholar] [CrossRef]
- Hirai, I.; Okuno, M.; Katsuma, R.; Arita, N.; Tachibana, M.; Yamamoto, Y. Characterisation of anti-Staphylococcus aureus activity of quercetin. Int. J. Food Sci. Technol. 2010, 45, 1250–1254. [Google Scholar] [CrossRef]
- Shu, Y.; Liu, Y.; Li, L.; Feng, J.; Lou, B.; Zhou, X.; Wu, H. Antibacterial activity of quercetin on oral infectious pathogens. Afr. J. Microbiol. Res. 2011, 5, 5358–5361. [Google Scholar]
- Tatsimo, S.J.N.; Tamokou, J.d.D.; Havyarimana, L.; Csupor, D.; Forgo, P.; Hohmann, J.; Kuiate, J.-R.; Tane, P. Antimicrobial and antioxidant activity of kaempferol rhamnoside derivatives from Bryophyllum pinnatum. BMC Res. Notes 2012, 5, 158. [Google Scholar] [CrossRef] [PubMed]
- Al-Rifai, A.A.A.; Ayoub, M.T.; Shakya, A.K.; Abu Safieh, K.A.; Mubarak, M.S. Synthesis, characterization, and antimicrobial activity of some new coumarin derivatives. Med. Chem. Res. 2012, 21, 468–476. [Google Scholar] [CrossRef]
- Wallock-Richards, D.; Doherty, C.J.; Doherty, L.; Clarke, D.J.; Place, M.; Govan, J.R.W.; Campopiano, D.J. Garlic Revisited: Antimicrobial Activity of Allicin-Containing Garlic Extracts against Burkholderia cepacia Complex. PLoS ONE 2014, 9, e112726. [Google Scholar] [CrossRef] [PubMed]
- Jaisinghani, R.N. Antibacterial properties of quercetin. Microbiol. Res. 2017, 8, 6877. [Google Scholar] [CrossRef]
- Di Sotto, A.; Di Giacomo, S.; Amatore, D.; Locatelli, M.; Vitalone, A.; Toniolo, C.; Rotino, G.; Lo Scalzo, R.; Palamara, A.; Marcocci, M.E.; et al. A Polyphenol Rich Extract from Solanum melongena L. DR2 Peel Exhibits Antioxidant Properties and Anti-Herpes Simplex Virus Type 1 Activity In Vitro. Molecules 2018, 23, 2066. [Google Scholar] [CrossRef]
- Di Sotto, A.; Checconi, P.; Celestino, I.; Locatelli, M.; Carissimi, S.; De Angelis, M.; Rossi, V.; Limongi, D.; Toniolo, C.; Martinoli, L.; et al. Antiviral and Antioxidant Activity of a Hydroalcoholic Extract from Humulus lupulus L. Oxidative Med. Cell. Longev. 2018, 2018, 5919237. [Google Scholar] [CrossRef]
- Nakamoto, M.; Kunimura, K.; Suzuki, J.I.; Kodera, Y. Antimicrobial properties of hydrophobic compounds in garlic: Allicin, vinyldithiin, ajoene and diallyl polysulfides (Review). Exp. Ther. Med. 2020, 19, 1550–1553. [Google Scholar] [CrossRef]
- Periferakis, A.; Periferakis, K.; Badarau, I.A.; Petran, E.M.; Popa, D.C.; Caruntu, A.; Costache, R.S.; Scheau, C.; Caruntu, C.; Costache, D.O. Kaempferol: Antimicrobial Properties, Sources, Clinical, and Traditional Applications. Int. J. Mol. Sci. 2022, 23, 15054. [Google Scholar] [CrossRef]
- Bangar, S.P.; Chaudhary, V.; Sharma, N.; Bansal, V.; Ozogul, F.; Lorenzo, J.M. Kaempferol: A flavonoid with wider biological activities and its applications. Crit. Rev. Food Sci. Nutr. 2022, 1–25. [Google Scholar] [CrossRef] [PubMed]
- Vuković, S.; Popović-Djordjević, J.B.; Kostić, A.Ž.; Pantelić, N.D.; Srećković, N.; Akram, M.; Laila, U.; Katanić Stanković, J.S. Allium Species in the Balkan Region–Major Metabolites, Antioxidant and Antimicrobial Properties. Horticulturae 2023, 9, 408. [Google Scholar] [CrossRef]
- Valipour, M.; Hosseini, A.; Di Sotto, A.; Irannejad, H. Dual action anti-inflammatory/antiviral isoquinoline alkaloids as potent naturally occurring anti-SARS-CoV-2 agents: A combined pharmacological and medicinal chemistry perspective. Phytother. Res. 2023, 37, 2168–2186. [Google Scholar] [CrossRef] [PubMed]
- Cordell, G.A.; Araujo, O.E. Capsaicin: Identification, nomenclature, and pharmacotherapy. Ann. Pharmacother. 1993, 27, 330–336. [Google Scholar] [CrossRef]
- Srinivasan, K. Biological Activities of Red Pepper (Capsicum annuum) and Its Pungent Principle Capsaicin: A Review. Crit. Rev. Food Sci. Nutr. 2016, 56, 1488–1500. [Google Scholar] [CrossRef] [PubMed]
- Naves, E.R.; de Ávila Silva, L.; Sulpice, R.; Araújo, W.L.; Nunes-Nesi, A.; Peres, L.E.P.; Zsögön, A. Capsaicinoids: Pungency beyond Capsicum. Trends Plant Sci. 2019, 24, 109–120. [Google Scholar] [CrossRef]
- Gradinaru, T.C.; Petran, M.; Dragos, D.; Gilca, M. PlantMolecularTasteDB: A Database of Taste Active Phytochemicals. Front. Pharmacol. 2021, 12, 751712. [Google Scholar] [CrossRef]
- Alberti, A.; Galasso, V.; Kovac, B.; Modelli, A.; Pichierri, F. Probing the molecular and electronic structure of capsaicin: A spectroscopic and quantum mechanical study. J. Phys. Chem. A 2008, 112, 5700–5711. [Google Scholar] [CrossRef]
- Reyes-Escogido Mde, L.; Gonzalez-Mondragon, E.G.; Vazquez-Tzompantzi, E. Chemical and pharmacological aspects of capsaicin. Molecules 2011, 16, 1253–1270. [Google Scholar] [CrossRef] [PubMed]
- Bennett, D.J.; Kirby, G.W. Constitution and biosynthesis of capsaicin. J. Chem. Soc. C Org. 1968, 442–446. [Google Scholar] [CrossRef]
- Leete, E.; Louden, M.C.L. Biosynthesis of capsaicin and dihydrocapsaicin in Capsicum frutescens. J. Am. Chem. Soc. 1968, 90, 6837–6841. [Google Scholar] [CrossRef]
- Aza-González, C.; Núñez-Palenius, H.G.; Ochoa-Alejo, N. Molecular biology of capsaicinoid biosynthesis in chili pepper (Capsicum spp.). Plant Cell Rep. 2011, 30, 695–706. [Google Scholar] [CrossRef] [PubMed]
- Kaiser, M.; Higuera, I.; Goycoolea, F.M. Capsaicinoids: Occurrence, Chemistry, Biosynthesis, and Biological Effects. In Fruit and Vegetable Phytochemicals; Wiley: Hoboken, NJ, USA, 2017; pp. 499–514. [Google Scholar] [CrossRef]
- Katsuragi, H.; Shimoda, K.; Yamamoto, R.; Ohara, T.; Hamada, H. Enzymatic synthesis of capsaicin 4-O-β-xylooligosaccharides by β-xylosidase from Aspergillus sp. Acta. Biol. Hung. 2011, 62, 151–155. [Google Scholar] [CrossRef]
- Cowles, R.S.; Keller, J.E.; Miller, J.R. Pungent spices, ground red pepper, and synthetic capsaicin as onion fly ovipositional deterrents. J. Chem. Ecol. 1989, 15, 719–730. [Google Scholar] [CrossRef] [PubMed]
- Kimball, B.A.; Taylor, J.; Perry, K.R.; Capelli, C. Deer responses to repellent stimuli. J. Chem. Ecol. 2009, 35, 1461–1470. [Google Scholar] [CrossRef]
- Stock, B.; Haag-Wackernagel, D. Effectiveness of Gel Repellents on Feral Pigeons. Animals 2013, 4, 1–15. [Google Scholar] [CrossRef]
- Yang, N.; Galves, C.; Racioni Goncalves, A.C.; Chen, J.; Fisk, I. Impact of capsaicin on aroma release: In vitro and in vivo analysis. Food Res. Int. 2020, 133, 109197. [Google Scholar] [CrossRef]
- Hu, X.; Ayed, C.; Chen, J.; Fisk, I.; Yang, N. The role of capsaicin stimulation on the physicochemical properties of saliva and aroma release in model aqueous and oil systems. Food Chem. 2022, 386, 132824. [Google Scholar] [CrossRef]
- Maliszewska, J.; Wyszkowska, J.; Kletkiewicz, H.; Rogalska, J. Capsaicin-induced dysregulation of acid-base status in the American cockroach. J. Environ. Sci. Health B 2019, 54, 676–680. [Google Scholar] [CrossRef] [PubMed]
- Cui, S.-F.; Wang, J.-W.; Li, H.-F.; Fang, R.; Yu, X.; Lu, Y.-J. Microencapsulation of Capsaicin in Chitosan Microcapsules: Characterization, Release Behavior, and Pesticidal Properties against Tribolium castaneum (Herbst). Insects 2023, 14, 27. [Google Scholar] [CrossRef]
- Adaszek, Ł.; Gadomska, D.; Mazurek, Ł.; Łyp, P.; Madany, J.; Winiarczyk, S. Properties of capsaicin and its utility in veterinary and human medicine. Res. Vet. Sci. 2019, 123, 14–19. [Google Scholar] [CrossRef]
- Mohammad, S.; Mustofa Helmi, E.; Faisal, F.; Muhammad Thohawi Elziyad, P. Role of Capsaicin in the Repair of Cellular Activity in Mice Liver. Pharmacogn. J. 2021, 13, 1573–1576. [Google Scholar]
- Watson, P.N.C.; Evans, R.J. The postmastectomy pain syndrome and topical capsaicin: A randomized trial. Pain 1992, 51, 375–379. [Google Scholar] [CrossRef]
- McCleane, G. The analgesic efficacy of topical capsaicin is enhanced by glyceryl trinitrate in painful osteoarthritis: A randomized, double blind, placebo controlled study. Eur. J. Pain 2000, 4, 355–360. [Google Scholar] [CrossRef] [PubMed]
- Grushka, M.; Epstein, J.B.; Gorsky, M. Burning mouth syndrome. Am. Fam. Physician 2002, 65, 615–620. [Google Scholar] [PubMed]
- Saguil, A.; Kane, S.; Mercado, M.; Lauters, R. Herpes Zoster and Postherpetic Neuralgia: Prevention and Management. Am. Fam. Physician 2017, 96, 656–663. [Google Scholar]
- Breneman, D.L.; Cardone, J.S.; Blumsack, R.F.; Lather, R.M.; Searle, E.A.; Pollack, V.E. Topical capsaicin for treatment of hemodialysis-related pruritus. J. Am. Acad. Dermatol. 1992, 26, 91–94. [Google Scholar] [CrossRef]
- Lysy, J.; Sistiery-Ittah, M.; Israelit, Y.; Shmueli, A.; Strauss-Liviatan, N.; Mindrul, V.; Keret, D.; Goldin, E. Topical capsaicin—A novel and effective treatment for idiopathic intractable pruritus ani: A randomised, placebo controlled, crossover study. Gut 2003, 52, 1323–1326. [Google Scholar] [CrossRef]
- Makhlough, A.; Ala, S.; Haj-Heydari, Z.; Kashi, Z.; Bari, A. Topical capsaicin therapy for uremic pruritus in patients on hemodialysis. Iran J. Kidney Dis. 2010, 4, 137–140. [Google Scholar]
- Gooding, S.M.; Canter, P.H.; Coelho, H.F.; Boddy, K.; Ernst, E. Systematic review of topical capsaicin in the treatment of pruritus. Int. J. Dermatol. 2010, 49, 858–865. [Google Scholar] [CrossRef]
- de Sèze, M.; Wiart, L.; de Sèze, M.P.; Soyeur, L.; Dosque, J.P.; Blajezewski, S.; Moore, N.; Brochet, B.; Mazaux, J.M.; Barat, M.; et al. Intravesical capsaicin versus resiniferatoxin for the treatment of detrusor hyperreflexia in spinal cord injured patients: A double-blind, randomized, controlled study. J. Urol. 2004, 171, 251–255. [Google Scholar] [CrossRef]
- Cruz, F. Mechanisms involved in new therapies for overactive bladder. Urology 2004, 63, 65–73. [Google Scholar] [CrossRef] [PubMed]
- Misra, M.N.; Pullani, A.J.; Mohamed, Z.U. Prevention of PONV by acustimulation with Capsicum plaster is comparable to ondansetron after middle ear surgery. Can. J. Anaesth 2005, 52, 485–489. [Google Scholar] [CrossRef] [PubMed]
- Agarwal, A.; Dhiraaj, S.; Tandon, M.; Singh, P.K.; Singh, U.; Pawar, S. Evaluation of capsaicin ointment at the Korean hand acupressure point K-D2 for prevention of postoperative nausea and vomiting. Anaesthesia 2005, 60, 1185–1188. [Google Scholar] [CrossRef] [PubMed]
- Hayman, M.; Kam, P.C.A. Capsaicin: A review of its pharmacology and clinical applications. Curr. Anaesth. Crit. Care 2008, 19, 338–343. [Google Scholar] [CrossRef]
- Botonis, P.G.; Miliotis, P.G.; Kounalakis, S.N.; Koskolou, M.D.; Geladas, N.D. Effects of capsaicin application on the skin during resting exposure to temperate and warm conditions. Scand J. Med. Sci. Sports 2019, 29, 171–179. [Google Scholar] [CrossRef]
- Rothenberger, J.; Wittwer, M.; Tschumi, C.; Constantinescu, M.A.; Daigeler, A.; Olariu, R. Quantitative impact analysis of remote ischemic conditioning and capsaicin application on human skin microcirculation. Clin. Hemorheol. Microcirc. 2019, 71, 291–298. [Google Scholar] [CrossRef] [PubMed]
- Caruntu, C.; Negrei, C.; Ilie Ghita, M.; Caruntu, A.; Bădărău, A.I.; Ioan, B.; Boda, D.; Albu, A.; Brănişteanu, D. Capsaicin, a Hot Topic in Skin Pharmacology and Physiology. Inflammation 2015, 63, 487–491. [Google Scholar]
- Ilie Ghita, M.; Caruntu, C.; Tampa, M.; Georgescu, S.R.; Matei, C.; Negrei, C.; Ion, R.-M.; Constantin, C.; Neagu, M.; Boda, D. Capsaicin: Physicochemical properties, cutaneous reactions and potential applications in painful and inflammatory conditions (Review). Exp. Ther. Med. 2019, 18, 916–925. [Google Scholar]
- Căruntu, C.; Boda, D. Evaluation through in vivo reflectance confocal microscopy of the cutaneous neurogenic inflammatory reaction induced by capsaicin in human subjects. J. Biomed. Opt. 2012, 17, 085003. [Google Scholar] [CrossRef] [PubMed]
- Lakatos, S.; Jancsó, G.; Horváth, Á.; Dobos, I.; Sántha, P. Longitudinal Study of Functional Reinnervation of the Denervated Skin by Collateral Sprouting of Peptidergic Nociceptive Nerves Utilizing Laser Doppler Imaging. Front. Physiol. 2020, 11, 439. [Google Scholar] [CrossRef] [PubMed]
- Wallace, R.J. Antimicrobial properties of plant secondary metabolites. Proc. Nutr. Soc. 2004, 63, 621–629. [Google Scholar] [CrossRef] [PubMed]
- Salehi, B.; Zakaria, Z.A.; Gyawali, R.; Ibrahim, S.A.; Rajkovic, J.; Shinwari, Z.K.; Khan, T.; Sharifi-Rad, J.; Ozleyen, A.; Turkdonmez, E.; et al. Piper Species: A Comprehensive Review on Their Phytochemistry, Biological Activities and Applications. Molecules 2019, 24, 1364. [Google Scholar] [CrossRef]
- Makuwa, S.C.; Serepa-Dlamini, M.H. The Antibacterial Activity of Crude Extracts of Secondary Metabolites from Bacterial Endophytes Associated with Dicoma anomala. Int. J. Microbiol. 2021, 2021, 8812043. [Google Scholar] [CrossRef] [PubMed]
- Aminah, N.S.; Laili, E.R.; Rafi, M.; Rochman, A.; Insanu, M.; Tun, K.N.W. Secondary metabolite compounds from Sida genus and their bioactivity. Heliyon 2021, 7, e06682. [Google Scholar] [CrossRef] [PubMed]
- Jubair, N.; Rajagopal, M.; Chinnappan, S.; Abdullah, N.B.; Fatima, A. Review on the Antibacterial Mechanism of Plant-Derived Compounds against Multidrug-Resistant Bacteria (MDR). Evid. Based Complement Altern. Med. 2021, 2021, 3663315. [Google Scholar] [CrossRef]
- Bhatia, P.; Sharma, A.; George, A.J.; Anvitha, D.; Kumar, P.; Dwivedi, V.P.; Chandra, N.S. Antibacterial activity of medicinal plants against ESKAPE: An update. Heliyon 2021, 7, e06310. [Google Scholar] [CrossRef]
- Füchtbauer, S.; Mousavi, S.; Bereswill, S.; Heimesaat, M.M. Antibacterial properties of capsaicin and its derivatives and their potential to fight antibiotic resistance—A literature survey. Eur. J. Microbiol. Immunol. 2021, 11, 10–17. [Google Scholar] [CrossRef]
- Kitsos, N.; Cassimos, D.; Xinias, I.; Agakidis, C.; Mavroudi, A. Management of antibiotic allergy in children: A practical approach. Allergol. Immunopathol. 2022, 50, 30–38. [Google Scholar] [CrossRef]
- Green, E.A.; Fogarty, K.; Ishmael, F.T. Penicillin Allergy: Mechanisms, Diagnosis, and Management. Prim. Care 2023, 50, 221–235. [Google Scholar] [CrossRef] [PubMed]
- Wrynn, A.F. An overview of penicillin allergies for nurses. Nursing 2023, 53, 27–31. [Google Scholar] [CrossRef] [PubMed]
- Westphal, J.; Vetter, D.; Brogard, J. Hepatic side-effects of antibiotics. J. Antimicrob. Chemother. 1994, 33, 387–401. [Google Scholar] [CrossRef] [PubMed]
- Thiim, M.; Friedman, L.S. Hepatotoxicity of antibiotics and antifungals. Clin. Liver Dis. 2003, 7, 381–399. [Google Scholar] [CrossRef]
- Polson, J.E. Hepatotoxicity due to antibiotics. Clin. Liver Dis. 2007, 11, 549–561. [Google Scholar] [CrossRef] [PubMed]
- Andrade, R.J.; Tulkens, P.M. Hepatic safety of antibiotics used in primary care. J. Antimicrob. Chemother. 2011, 66, 1431–1446. [Google Scholar] [CrossRef] [PubMed]
- Rehman, K.; Kamran, S.H.; Hamid Akash, M.S. Chapter 16—Toxicity of antibiotics. In Antibiotics and Antimicrobial Resistance Genes in the Environment; Hashmi, M.Z., Ed.; Elsevier: Amsterdam, The Netherlands, 2020; Volume 1, pp. 234–252. [Google Scholar]
- Dorobăţ, O.-M.; Moisoiu, A.; Tălăpan, D. Bacteria isolated from pleural fluid and their resistance to antimicrobials. Pneumologia 2006, 55, 47–51. [Google Scholar] [PubMed]
- Dorobăţ, O.M.; Moisoiu, A.; Tălăpan, D. Incidence and resistance patterns of pathogens from lower respiratory tract infections (LRTI). Pneumologia 2007, 56, 7–15. [Google Scholar]
- Dorobăţ, O.M.; Bădicuţ, I.; Tălăpan, D.; Tenea, C.; Rafila, A. Antibiotic resistance of Gram-positive cocci isolated in 2008. Bacteriol. Virusol. Parazitol. Epidemiol. 2010, 55, 83–92. [Google Scholar]
- Popescu, G.A.; Șerban, R.; Iosif, I.; Codiță, I.; Dorobăț, O.; Tălăpan, D.; Buzea, M.; Szekely, E.; Dorneanu, O.; Bota, K.; et al. Antimicrobial resistance of germs isolated from invasive infections–Romania 2012. BMC Infect. Dis. 2013, 13, O16. [Google Scholar] [CrossRef]
- Rafila, A.; Talapan, D.; Dorobăţ, O.M.; Popescu, G.A.; Piţigoi, D.; Florea, D.; Buicu, F.C. Emergence of Carbapenemase-producing Enterobacteriaceae, a Public Health Threat: A Romanian Infectious Disease Hospital Based Study/Emergenţa Enterobacteriaceaelor producătoare de carbapenemaze, o ameninţare pentru sănătatea publică: Un studiu realizat într-un spital romanesc de boli infectioase. Rev. Romana Med. Lab. 2015, 23, 295–301. [Google Scholar] [CrossRef]
- Kohler, P.P.; Melano, R.G.; Patel, S.N.; Shafinaz, S.; Faheem, A.; Coleman, B.L.; Green, K.; Armstrong, I.; Almohri, H.; Borgia, S.; et al. Emergence of Carbapenemase-Producing Enterobacteriaceae, South-Central Ontario, Canada. Emerg. Infect. Dis. 2018, 24, 1674–1682. [Google Scholar] [CrossRef] [PubMed]
- Tălăpan, D.; Rafila, A. Five-Year Survey of Asymptomatic Colonization with Multidrug-Resistant Organisms in a Romanian Tertiary Care Hospital. Infect. Drug Resist. 2022, 15, 2959–2967. [Google Scholar] [CrossRef] [PubMed]
- Ackerman, S.; Gonzales, R. The context of antibiotic overuse. Ann. Intern. Med. 2012, 157, 211–212. [Google Scholar] [CrossRef] [PubMed]
- Shallcross, L.J.; Davies, D.S.C. Antibiotic overuse: A key driver of antimicrobial resistance. Br. J. Gen. Pract. 2014, 64, 604–605. [Google Scholar] [CrossRef]
- Willis, L.D.; Chandler, C. Quick fix for care, productivity, hygiene and inequality: Reframing the entrenched problem of antibiotic overuse. BMJ Glob. Health 2019, 4, e001590. [Google Scholar] [CrossRef] [PubMed]
- Kanellopoulos, C.; Lamprinou, V.; Politi, A.; Voudouris, P.; Iliopoulos, I.; Kokkaliari, M.; Moforis, L.; Economou-Amilli, A. Microbial Mat Stratification in Travertine Depositions of Greek Hot Springs and Biomineralization Processes. Minerals 2022, 12, 1408. [Google Scholar] [CrossRef]
- Kanellopoulos, C.; Lamprinou, V.; Politi, A.; Voudouris, P.; Iliopoulos, I.; Kokkaliari, M.; Moforis, L.; Economou-Amilli, A. Speleothems and Biomineralization Processes in Hot Spring Environment: The Case of Aedipsos (Edipsos), Euboea (Evia) Island, Greece. J. Mar. Sci. Eng. 2022, 10, 1909. [Google Scholar] [CrossRef]
- Kanellopoulos, C.; Lamprinou, V.; Politi, A.; Voudouris, P.; Economou-Amilli, A. Pioneer species of Cyanobacteria in hot springs and their role to travertine formation: The case of Aedipsos hot springs, Euboea (Evia), Greece. Depos. Rec. 2022, 8, 1079–1092. [Google Scholar] [CrossRef]
- EUCAST. Definitive Document Methods for the determination of susceptibility of bacteria to antimicrobial agents. Terminology. Clin Microbiol Infect 1998, 4, 291. [Google Scholar]
- Nascimento, P.L.; Nascimento, T.C.; Ramos, N.S.; Silva, G.R.; Gomes, J.E.; Falcão, R.E.; Moreira, K.A.; Porto, A.L.; Silva, T.M. Quantification, antioxidant and antimicrobial activity of phenolics isolated from different extracts of Capsicum frutescens (Pimenta Malagueta). Molecules 2014, 19, 5434–5447. [Google Scholar] [CrossRef]
- Das, J.; Deka, M.; Gogoi, K. Antimicrobial activity of chilli extracts (Capsicum chinense) against food borne pathogens Escherichia coli and Staphylococcus aureus. Int. J. Res. Anal. Rev. 2018, 5, 717–720. [Google Scholar]
- Marini, E.; Magi, G.; Mingoia, M.; Pugnaloni, A.; Facinelli, B. Antimicrobial and Anti-Virulence Activity of Capsaicin against Erythromycin-Resistant, Cell-Invasive Group A Streptococci. Front. Microbiol. 2015, 6, 1281. [Google Scholar] [CrossRef]
- Omolo, M.A.; Wong, Z.-Z.; Borh, W.G.; Hedblom, G.A.; Dev, K.; Baumler, D.J. Comparative analysis of capsaicin in twenty nine varieties of unexplored Capsicum and its antimicrobial activity against bacterial and fungal pathogens. J. Med. Plants Res. 2018, 12, 544–556. [Google Scholar]
- Chatterjee, S.; Asakura, M.; Chowdhury, N.; Neogi, S.B.; Sugimoto, N.; Haldar, S.; Awasthi, S.P.; Hinenoya, A.; Aoki, S.; Yamasaki, S. Capsaicin, a potential inhibitor of cholera toxin production in Vibrio cholerae. FEMS Microbiol. Lett 2010, 306, 54–60. [Google Scholar] [CrossRef] [PubMed]
- Ozçelik, B.; Kartal, M.; Orhan, I. Cytotoxicity, antiviral and antimicrobial activities of alkaloids, flavonoids, and phenolic acids. Pharm. Biol. 2011, 49, 396–402. [Google Scholar] [CrossRef] [PubMed]
- Zeyrek, F.Y.; Oguz, E. In vitro activity of capsaicin against Helicobacter pylori. Ann. Microbiol. 2005, 55, 125–127. [Google Scholar]
- Ayariga, J.A.; Abugri, D.A.; Amrutha, B.; Villafane, R. Capsaicin Potently Blocks Salmonella typhimurium Invasion of Vero Cells. Antibiotics 2022, 11, 666. [Google Scholar] [CrossRef]
- Kushwaha, M.; Jain, S.K.; Sharma, N.; Abrol, V.; Jaglan, S.; Vishwakarma, R.A. Establishment of LCMS Based Platform for Discovery of Quorum Sensing Inhibitors: Signal Detection in Pseudomonas aeruginosa PAO1. ACS Chem. Biol. 2018, 13, 657–665. [Google Scholar] [CrossRef]
- Ahmad-Mansour, N.; Loubet, P.; Pouget, C.; Dunyach-Remy, C.; Sotto, A.; Lavigne, J.P.; Molle, V. Staphylococcus aureus Toxins: An Update on Their Pathogenic Properties and Potential Treatments. Toxins 2021, 13, 677. [Google Scholar] [CrossRef] [PubMed]
- Leung, A.K.C.; Barankin, B.; Leong, K.F. Staphylococcal-scalded skin syndrome: Evaluation, diagnosis, and management. World J. Pediatr. 2018, 14, 116–120. [Google Scholar] [CrossRef] [PubMed]
- Lowy, F.D. Staphylococcus aureus infections. N. Engl. J. Med. 1998, 339, 520–532. [Google Scholar] [CrossRef] [PubMed]
- Tong, S.Y.; Davis, J.S.; Eichenberger, E.; Holland, T.L.; Fowler, V.G., Jr. Staphylococcus aureus infections: Epidemiology, pathophysiology, clinical manifestations, and management. Clin. Microbiol. Rev. 2015, 28, 603–661. [Google Scholar] [CrossRef]
- Stevens, D.L. The toxins of group A streptococcus, the flesh eating bacteria. Immunol. Investig. 1997, 26, 129–150. [Google Scholar] [CrossRef]
- Malachowa, N.; DeLeo, F.R. Mobile genetic elements of Staphylococcus aureus. Cell Mol. Life Sci. 2010, 67, 3057–3071. [Google Scholar] [CrossRef]
- Howden, B.P.; Davies, J.K.; Johnson, P.D.; Stinear, T.P.; Grayson, M.L. Reduced vancomycin susceptibility in Staphylococcus aureus, including vancomycin-intermediate and heterogeneous vancomycin-intermediate strains: Resistance mechanisms, laboratory detection, and clinical implications. Clin. Microbiol. Rev. 2010, 23, 99–139. [Google Scholar] [CrossRef]
- Grema, H.A.; Geidam, Y.A.; Gadzama, G.B.; Ameh, J.A.; Suleiman, A. Methicillin resistant Staphylococcus aureus (MRSA): A review. Adv. Anim. Vet. Sci. 2015, 3, 79–98. [Google Scholar] [CrossRef]
- Akyuz, L.; Kaya, M.; Mujtaba, M.; Ilk, S.; Sargin, I.; Salaberria, A.M.; Labidi, J.; Cakmak, Y.S.; Islek, C. Supplementing capsaicin with chitosan-based films enhanced the anti-quorum sensing, antimicrobial, antioxidant, transparency, elasticity and hydrophobicity. Int. J. Biol. Macromol. 2018, 115, 438–446. [Google Scholar] [CrossRef]
- Qiu, J.; Niu, X.; Wang, J.; Xing, Y.; Leng, B.; Dong, J.; Li, H.; Luo, M.; Zhang, Y.; Dai, X. Capsaicin protects mice from community-associated methicillin-resistant Staphylococcus aureus pneumonia. PLoS ONE 2012, 7, e33032. [Google Scholar] [CrossRef]
- Cunningham, M.W. Pathogenesis of group A streptococcal infections. Clin. Microbiol. Rev. 2000, 13, 470–511. [Google Scholar] [CrossRef]
- Jespersen, M.G.; Lacey, J.A.; Tong, S.Y.C.; Davies, M.R. Global genomic epidemiology of Streptococcus pyogenes. Infect. Genet. Evol. 2020, 86, 104609. [Google Scholar] [CrossRef]
- Lynskey, N.N.; Lawrenson, R.A.; Sriskandan, S. New understandings in Streptococcus pyogenes. Curr. Opin. Infect. Dis. 2011, 24, 196–202. [Google Scholar] [CrossRef] [PubMed]
- González-Abad, M.J.; Alonso Sanz, M. Infecciones invasoras por Streptococcus pyogenes (2011–2018): Serotipos y presentación clínica. An. Pediatría 2020, 92, 351–358. [Google Scholar] [CrossRef] [PubMed]
- Sriskandan, S.; Faulkner, L.; Hopkins, P. Streptococcus pyogenes: Insight into the function of the streptococcal superantigens. Int. J. Biochem. Cell Biol. 2007, 39, 12–19. [Google Scholar] [CrossRef]
- Alouf, J.E. Streptococcal toxins (streptolysin O, streptolysin S, erythrogenic toxin). Pharmacol. Ther. 1980, 11, 661–717. [Google Scholar] [CrossRef]
- Noskin, G.A.; Peterson, L.R.; Warren, J.R. Enterococcus faecium and Enterococcus faecalis bacteremia: Acquisition and outcome. Clin. Infect. Dis. 1995, 20, 296–301. [Google Scholar] [CrossRef]
- Schaberg, D.R.; Culver, D.H.; Gaynes, R.P. Major trends in the microbial etiology of nosocomial infection. Am. J. Med. 1991, 91, S72–S75. [Google Scholar] [CrossRef] [PubMed]
- Kaye, D. Enterococci: Biologic and epidemiologic characteristics and in vitro susceptibility. Arch. Intern. Med. 1982, 142, 2006–2009. [Google Scholar] [CrossRef]
- Maki, D.G.; Agger, W.A. Enterococcal Bacteremia: Clinical Features, the Risk of Endocarditis, and Management. Medicine 1988, 67, 248. [Google Scholar] [CrossRef] [PubMed]
- Cetinkaya, Y.; Falk, P.; Mayhall, C.G. Vancomycin-resistant enterococci. Clin. Microbiol. Rev. 2000, 13, 686–707. [Google Scholar] [CrossRef]
- Flagan, S.F.; Leadbetter, J.R. Utilization of capsaicin and vanillylamine as growth substrates by Capsicum (hot pepper)-associated bacteria. Environ. Microbiol. 2006, 8, 560–565. [Google Scholar] [CrossRef] [PubMed]
- Gu, H.J.; Sun, Q.L.; Luo, J.C.; Zhang, J.; Sun, L. A First Study of the Virulence Potential of a Bacillus subtilis Isolate from Deep-Sea Hydrothermal Vent. Front. Cell Infect. Microbiol. 2019, 9, 183. [Google Scholar] [CrossRef]
- Brown, K.L. Control of bacterial spores. Br. Med. Bull. 2000, 56, 158–171. [Google Scholar] [CrossRef] [PubMed]
- Cote, C.K.; Heffron, J.D.; Bozue, J.A.; Welkos, S.L. Chapter 102—Bacillus anthracis and Other Bacillus Species. In Molecular Medical Microbiology, 2nd ed.; Tang, Y.-W., Sussman, M., Liu, D., Poxton, I., Schwartzman, J., Eds.; Academic Press: Boston, MA, USA, 2015; pp. 1789–1844. [Google Scholar] [CrossRef]
- Spencer, R.C. Bacillus anthracis. J. Clin. Pathol. 2003, 56, 182–187. [Google Scholar] [CrossRef]
- Molina-Torres, J.; García-Chávez, A.; Ramírez-Chávez, E. Antimicrobial properties of alkamides present in flavouring plants traditionally used in Mesoamerica: Affinin and capsaicin. J. Ethnopharmacol. 1999, 64, 241–248. [Google Scholar] [CrossRef]
- Argôlo-Filho, R.C.; Loguercio, L.L. Bacillus thuringiensis Is an Environmental Pathogen and Host-Specificity Has Developed as an Adaptation to Human-Generated Ecological Niches. Insects 2013, 5, 62–91. [Google Scholar] [CrossRef] [PubMed]
- Farber, J.M.; Peterkin, P.I. Listeria monocytogenes, a food-borne pathogen. Microbiol. Rev. 1991, 55, 476–511. [Google Scholar] [CrossRef]
- Schwarzkopf, A. Listeria monocytogenes–aspects of pathogenicity. Pathol. Biol. 1996, 44, 769–774. [Google Scholar]
- Disson, O.; Moura, A.; Lecuit, M. Making Sense of the Biodiversity and Virulence of Listeria monocytogenes. Trends Microbiol. 2021, 29, 811–822. [Google Scholar] [CrossRef]
- Colwell, R.R.; Kaper, J.; Joseph, S.W. Vibrio cholerae, Vibrio parahaemolyticus, and other vibrios: Occurrence and distribution in Chesapeake Bay. Science 1977, 198, 394–396. [Google Scholar]
- Garay, E.; Arnau, A.; Amaro, C. Incidence of Vibrio cholerae and related vibrios in a coastal lagoon and seawater influenced by lake discharges along an annual cycle. Appl. Environ. Microbiol. 1985, 50, 426–430. [Google Scholar] [CrossRef] [PubMed]
- Reidl, J.; Klose, K.E. Vibrio cholerae and cholera: Out of the water and into the host. FEMS Microbiol. Rev. 2002, 26, 125–139. [Google Scholar] [CrossRef] [PubMed]
- Glass, R.I.; Huq, I.; Alim, A.R.; Yunus, M. Emergence of multiply antibiotic-resistant Vibrio cholerae in Bangladesh. J. Infect. Dis. 1980, 142, 939–942. [Google Scholar] [CrossRef]
- Das, B.; Verma, J.; Kumar, P.; Ghosh, A.; Ramamurthy, T. Antibiotic resistance in Vibrio cholerae: Understanding the ecology of resistance genes and mechanisms. Vaccine 2020, 38 (Suppl. S1), A83–A92. [Google Scholar] [CrossRef]
- Alam, M.; Islam, M.T.; Rashed, S.M.; Johura, F.T.; Bhuiyan, N.A.; Delgado, G.; Morales, R.; Mendez, J.L.; Navarro, A.; Watanabe, H.; et al. Vibrio cholerae classical biotype strains reveal distinct signatures in Mexico. J. Clin. Microbiol. 2012, 50, 2212–2216. [Google Scholar] [CrossRef]
- Asif, M.; Alvi, I.A.; Rehman, S.U. Insight into Acinetobacter baumannii: Pathogenesis, global resistance, mechanisms of resistance, treatment options, and alternative modalities. Infect. Drug Resist. 2018, 11, 1249–1260. [Google Scholar] [CrossRef] [PubMed]
- Pourhajibagher, M.; Hashemi, F.B.; Pourakbari, B.; Aziemzadeh, M.; Bahador, A. Antimicrobial Resistance of Acinetobacter baumannii to Imipenem in Iran: A Systematic Review and Meta-Analysis. Open Microbiol. J. 2016, 10, 32–42. [Google Scholar] [CrossRef] [PubMed]
- Qi, L.; Li, H.; Zhang, C.; Liang, B.; Li, J.; Wang, L.; Du, X.; Liu, X.; Qiu, S.; Song, H. Relationship between Antibiotic Resistance, Biofilm Formation, and Biofilm-Specific Resistance in Acinetobacter baumannii. Front. Microbiol. 2016, 7, 483. [Google Scholar] [CrossRef]
- Gheorghe-Barbu, I.; Barbu, I.C.; Popa, L.I.; Pîrcălăbioru, G.G.; Popa, M.; Măruțescu, L.; Niță-Lazar, M.; Banciu, A.; Stoica, C.; Gheorghe, Ș.; et al. Temporo-spatial variations in resistance determinants and clonality of Acinetobacter baumannii and Pseudomonas aeruginosa strains from Romanian hospitals and wastewaters. Antimicrob. Resist. Infect. Control 2022, 11, 115. [Google Scholar] [CrossRef]
- Guo, T.; Li, M.; Sun, X.; Wang, Y.; Yang, L.; Jiao, H.; Li, G. Synergistic Activity of Capsaicin and Colistin against Colistin-Resistant Acinetobacter baumannii: In Vitro/Vivo Efficacy and Mode of Action. Front. Pharmacol. 2021, 12, 744494. [Google Scholar] [CrossRef] [PubMed]
- Mégraud, F. Resistance of Helicobacter pylori to antibiotics and its impact on treatment options. Drug Resist. Updat. 2001, 4, 178–186. [Google Scholar] [CrossRef]
- Dascălu, R.; Bolocan, A.; Păduaru, D.; Constantinescu, A.; Mitache, M.; Stoica, A.; Andronic, O. Multidrug resistance in Helicobacter pylori infection. Front. Microbiol. 2023, 14, 1128497. [Google Scholar] [CrossRef] [PubMed]
- Jones, N.L.; Shabib, S.; Sherman, P.M. Capsaicin as an inhibitor of the growth of the gastric pathogen Helicobacter pylori. FEMS Microbiol. Lett. 1997, 146, 223–227. [Google Scholar] [CrossRef]
- Saha, K.; Sarkar, D.; Khan, U.; Karmakar, B.C.; Paul, S.; Mukhopadhyay, A.K.; Dutta, S.; Bhattacharya, S. Capsaicin Inhibits Inflammation and Gastric Damage during H pylori Infection by Targeting NF-kB-miRNA Axis. Pathogens 2022, 11, 641. [Google Scholar] [CrossRef] [PubMed]
- Venier, N.A.; Colquhoun, A.J.; Sasaki, H.; Kiss, A.; Sugar, L.; Adomat, H.; Fleshner, N.E.; Klotz, L.H.; Venkateswaran, V. Capsaicin: A novel radio-sensitizing agent for prostate cancer. Prostate 2015, 75, 113–125. [Google Scholar] [CrossRef]
- Crump, J.A.; Luby, S.P.; Mintz, E.D. The global burden of typhoid fever. Bull. World Health Organ. 2004, 82, 346–353. [Google Scholar]
- Popa, G.L.; Papa, M.I. Salmonella spp. infection—A continuous threat worldwide. Germs 2021, 11, 88–96. [Google Scholar] [CrossRef]
- Allerberger, F.; Liesegang, A.; Grif, K.; Khaschabi, D.; Prager, R.; Danzl, J.; Höck, F.; Ottl, J.; Dierich, M.P.; Berghold, C.; et al. Occurrence of Salmonella enterica serovar Dublin in Austria. Wien Med. Wochenschr. 2003, 153, 148–152. [Google Scholar] [CrossRef]
- Elias, A.; Viana, J.X.; Rangel, H.; Osles, A.G. Antigenic variation in Salmonella typhimurium. Proc. Soc. Exp. Biol. Med. 1974, 145, 392–396. [Google Scholar] [CrossRef]
- Martin, L.J.; Fyfe, M.; Doré, K.; Buxton, J.A.; Pollari, F.; Henry, B.; Middleton, D.; Ahmed, R.; Jamieson, F.; Ciebin, B.; et al. Increased Burden of Illness Associated with Antimicrobial-Resistant Salmonella enterica Serotype Typhimurium Infections. J. Infect. Dis. 2004, 189, 377–384. [Google Scholar] [CrossRef] [PubMed]
- Hussain, A.; Satti, L.; Hanif, F.; Zehra, N.M.; Nadeem, S.; Bangash, T.M.; Peter, A. Typhoidal Salmonella strains in Pakistan: An impending threat of extensively drug-resistant Salmonella Typhi. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 2145–2149. [Google Scholar] [CrossRef] [PubMed]
- Butaye, P.; Michael, G.B.; Schwarz, S.; Barrett, T.J.; Brisabois, A.; White, D.G. The clonal spread of multidrug-resistant non-typhi Salmonella serotypes. Microbes Infect. 2006, 8, 1891–1897. [Google Scholar] [CrossRef] [PubMed]
- Chiu, C.H.; Wu, T.L.; Su, L.H.; Chu, C.; Chia, J.H.; Kuo, A.J.; Chien, M.S.; Lin, T.Y. The emergence in Taiwan of fluoroquinolone resistance in Salmonella enterica serotype choleraesuis. N. Engl. J. Med. 2002, 346, 413–419. [Google Scholar] [CrossRef]
- Kaper, J.B.; Nataro, J.P.; Mobley, H.L. Pathogenic Escherichia coli. Nat. Rev. Microbiol. 2004, 2, 123–140. [Google Scholar] [CrossRef]
- Gomes, T.A.; Elias, W.P.; Scaletsky, I.C.; Guth, B.E.; Rodrigues, J.F.; Piazza, R.M.; Ferreira, L.C.; Martinez, M.B. Diarrheagenic Escherichia coli. Braz. J. Microbiol. 2016, 47 (Suppl. S1), 3–30. [Google Scholar] [CrossRef]
- Cerceo, E.; Deitelzweig, S.B.; Sherman, B.M.; Amin, A.N. Multidrug-Resistant Gram-Negative Bacterial Infections in the Hospital Setting: Overview, Implications for Clinical Practice, and Emerging Treatment Options. Microb. Drug Resist. 2016, 22, 412–431. [Google Scholar] [CrossRef]
- Zhang, Y.; Yang, J.; Ye, L.; Luo, Y.; Wang, W.; Zhou, W.; Cui, Z.; Han, L. Characterization of clinical multidrug-resistant Escherichia coli and Klebsiella pneumoniae isolates, 2007–2009, China. Microb. Drug Resist. 2012, 18, 465–470. [Google Scholar] [CrossRef]
- Navon-Venezia, S.; Kondratyeva, K.; Carattoli, A. Klebsiella pneumoniae: A major worldwide source and shuttle for antibiotic resistance. FEMS Microbiol. Rev. 2017, 41, 252–275. [Google Scholar] [CrossRef]
- Effah, C.Y.; Sun, T.; Liu, S.; Wu, Y. Klebsiella pneumoniae: An increasing threat to public health. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 1. [Google Scholar] [CrossRef]
- Podschun, R.; Ullmann, U. Klebsiella spp. as nosocomial pathogens: Epidemiology, taxonomy, typing methods, and pathogenicity factors. Clin. Microbiol. Rev. 1998, 11, 589–603. [Google Scholar] [CrossRef] [PubMed]
- Surleac, M.; Czobor Barbu, I.; Paraschiv, S.; Popa, L.I.; Gheorghe, I.; Marutescu, L.; Popa, M.; Sarbu, I.; Talapan, D.; Nita, M.; et al. Whole genome sequencing snapshot of multi-drug resistant Klebsiella pneumoniae strains from hospitals and receiving wastewater treatment plants in Southern Romania. PLoS ONE 2020, 15, e0228079. [Google Scholar] [CrossRef] [PubMed]
- Hassan, M.G.; Abdulrazik, G.; El Awady, M.E.; Hamed, A.A.; Abdel-Monem, M.O. Antimicrobial Activity of Capsaicin and Its Derivatives against Klebsiella pneumoniae. Egypt. Acad. J. Biol. Sci. G. Microbiol. 2021, 13, 79–90. [Google Scholar] [CrossRef]
- Al-Musawi, S.; Albukhaty, S.; Al-Karagoly, H.; Sulaiman, G.M.; Alwahibi, M.S.; Dewir, Y.H.; Soliman, D.A.; Rizwana, H. Antibacterial Activity of Honey/Chitosan Nanofibers Loaded with Capsaicin and Gold Nanoparticles for Wound Dressing. Molecules 2020, 25, 4770. [Google Scholar] [CrossRef]
- Papazafiropoulou, A.; Daniil, I.; Sotiropoulos, A.; Balampani, E.; Kokolaki, A.; Bousboulas, S.; Konstantopoulou, S.; Skliros, E.; Petropoulou, D.; Pappas, S. Prevalence of asymptomatic bacteriuria in type 2 diabetic subjects with and without microalbuminuria. BMC Res. Notes 2010, 3, 169. [Google Scholar] [CrossRef] [PubMed]
- Matthews, S.J.; Lancaster, J.W. Urinary tract infections in the elderly population. Am. J. Geriatr. Pharmacother. 2011, 9, 286–309. [Google Scholar] [CrossRef]
- Rodriguez-Mañas, L. Urinary tract infections in the elderly: A review of disease characteristics and current treatment options. Drugs Context 2020, 9, 2020-4-13. [Google Scholar] [CrossRef]
- Schaffer, J.N.; Pearson, M.M. Proteus mirabilis and Urinary Tract Infections. Microbiol. Spectr. 2015, 3. [Google Scholar] [CrossRef]
- Mobley, H.L.; Warren, J.W. Urease-positive bacteriuria and obstruction of long-term urinary catheters. J. Clin. Microbiol. 1987, 25, 2216–2217. [Google Scholar] [CrossRef]
- Mulvey, M.A.; Klumpp, D.J.; Stapleton, A.E. Urinary Tract Infections: Molecular Pathogenesis and Clinical Management; Wiley: Hoboken, NJ, USA, 2020. [Google Scholar]
- Hall, R.M.; Collis, C.M. Antibiotic resistance in gram-negative bacteria: The role of gene cassettes and integrons. Drug Resist. Updat. 1998, 1, 109–119. [Google Scholar] [CrossRef]
- Tumbarello, M.; Trecarichi, E.M.; Fiori, B.; Losito, A.R.; D’Inzeo, T.; Campana, L.; Ruggeri, A.; Di Meco, E.; Liberto, E.; Fadda, G.; et al. Multidrug-resistant Proteus mirabilis bloodstream infections: Risk factors and outcomes. Antimicrob. Agents Chemother. 2012, 56, 3224–3231. [Google Scholar] [CrossRef] [PubMed]
- Pagani, L.; Migliavacca, R.; Pallecchi, L.; Matti, C.; Giacobone, E.; Amicosante, G.; Romero, E.; Rossolini, G.M. Emerging extended-spectrum beta-lactamases in Proteus mirabilis. J. Clin. Microbiol. 2002, 40, 1549–1552. [Google Scholar] [CrossRef] [PubMed]
- Endimiani, A.; Luzzaro, F.; Brigante, G.; Perilli, M.; Lombardi, G.; Amicosante, G.; Rossolini, G.M.; Toniolo, A. Proteus mirabilis bloodstream infections: Risk factors and treatment outcome related to the expression of extended-spectrum beta-lactamases. Antimicrob. Agents Chemother. 2005, 49, 2598–2605. [Google Scholar] [CrossRef] [PubMed]
- Cohen-Nahum, K.; Saidel-Odes, L.; Riesenberg, K.; Schlaeffer, F.; Borer, A. Urinary tract infections caused by multi-drug resistant Proteus mirabilis: Risk factors and clinical outcomes. Infection 2010, 38, 41–46. [Google Scholar] [CrossRef] [PubMed]
- D’Andrea, M.M.; Literacka, E.; Zioga, A.; Giani, T.; Baraniak, A.; Fiett, J.; Sadowy, E.; Tassios, P.T.; Rossolini, G.M.; Gniadkowski, M.; et al. Evolution and spread of a multidrug-resistant Proteus mirabilis clone with chromosomal AmpC-type cephalosporinases in Europe. Antimicrob. Agents Chemother. 2011, 55, 2735–2742. [Google Scholar] [CrossRef]
- Charkhian, H.; Bodaqlouie, A.; Soleimannezhadbari, E.; Lotfollahi, L.; Shaykh-Baygloo, N.; Hosseinzadeh, R.; Yousefi, N.; Khodayar, M. Comparing the Bacteriostatic Effects of Different Metal Nanoparticles against Proteus vulgaris. Curr. Microbiol. 2020, 77, 2674–2684. [Google Scholar] [CrossRef]
- Klockgether, J.; Tümmler, B. Recent advances in understanding Pseudomonas aeruginosa as a pathogen. F1000Res 2017, 6, 1261. [Google Scholar] [CrossRef] [PubMed]
- Buhl, M.; Peter, S.; Willmann, M. Prevalence and risk factors associated with colonization and infection of extensively drug-resistant Pseudomonas aeruginosa: A systematic review. Expert Rev. Anti. Infect. Ther. 2015, 13, 1159–1170. [Google Scholar] [CrossRef] [PubMed]
- Murphy, T.F. Pseudomonas aeruginosa in adults with chronic obstructive pulmonary disease. Curr. Opin. Pulm. Med. 2009, 15, 138–142. [Google Scholar] [CrossRef]
- Gonçalves-de-Albuquerque, C.F.; Silva, A.R.; Burth, P.; Rocco, P.R.; Castro-Faria, M.V.; Castro-Faria-Neto, H.C. Possible mechanisms of Pseudomonas aeruginosa-associated lung disease. Int. J. Med. Microbiol. 2016, 306, 20–28. [Google Scholar] [CrossRef]
- Talwalkar, J.S.; Murray, T.S. The Approach to Pseudomonas aeruginosa in Cystic Fibrosis. Clin. Chest Med. 2016, 37, 69–81. [Google Scholar] [CrossRef]
- Hancock, R.E. Resistance mechanisms in Pseudomonas aeruginosa and other nonfermentative gram-negative bacteria. Clin. Infect. Dis. 1998, 27 (Suppl. S1), S93–S99. [Google Scholar] [CrossRef]
- Pang, Z.; Raudonis, R.; Glick, B.R.; Lin, T.J.; Cheng, Z. Antibiotic resistance in Pseudomonas aeruginosa: Mechanisms and alternative therapeutic strategies. Biotechnol. Adv. 2019, 37, 177–192. [Google Scholar] [CrossRef]
- Brown, G.D.; Denning, D.W.; Levitz, S.M. Tackling Human Fungal Infections. Science 2012, 336, 647. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Tudela, J.; Alastruey-Izquierdo, A.; Gago, S.; Cuenca-Estrella, M.; León, C.; Miro, J.; Boluda, A.N.; Camps, I.R.; Sole, A.; Denning, D. Burden of serious fungal infections in Spain. Clin. Microbiol. Infect. 2015, 21, 183–189. [Google Scholar] [CrossRef] [PubMed]
- Alvarez-Moreno, C.A.; Cortes, J.A.; Denning, D.W. Burden of fungal infections in Colombia. J. Fungi 2018, 4, 41. [Google Scholar] [CrossRef] [PubMed]
- Tufa, T.B.; Denning, D.W. The Burden of Fungal Infections in Ethiopia. J. Fungi 2019, 5, 109. [Google Scholar] [CrossRef]
- Fisher, M.C.; Hawkins, N.J.; Sanglard, D.; Gurr, S.J. Worldwide emergence of resistance to antifungal drugs challenges human health and food security. Science 2018, 360, 739–742. [Google Scholar] [CrossRef]
- Revie, N.M.; Iyer, K.R.; Robbins, N.; Cowen, L.E. Antifungal drug resistance: Evolution, mechanisms and impact. Curr. Opin. Microbiol. 2018, 45, 70–76. [Google Scholar] [CrossRef]
- Ben-Ami, R.; Kontoyiannis, D.P. Resistance to antifungal drugs. Infect. Dis. Clin. 2021, 35, 279–311. [Google Scholar] [CrossRef]
- Elewski, B.E. Mechanisms of action of systemic antifungal agents. J. Am. Acad. Dermatol. 1993, 28, S28–S34. [Google Scholar] [CrossRef] [PubMed]
- Dixon, D.M.; Walsh, T.J. Antifungal agents. In Medical Microbiology, 4th ed.; University of Texas Medical Branch at Galveston: Galveston, TX, USA, 1996. [Google Scholar]
- Gupta, A.K.; Tomas, E. New antifungal agents. Dermatol. Clin. 2003, 21, 565–576. [Google Scholar] [CrossRef]
- Menezes, R.P.; Bessa, M.A.S.; Siqueira, C.P.; Teixeira, S.C.; Ferro, E.A.V.; Martins, M.M.; Cunha, L.C.S.; Martins, C.H.G. Antimicrobial, Antivirulence, and Antiparasitic Potential of Capsicum chinense Jacq. Extracts and Their Isolated Compound Capsaicin. Antibiotics 2022, 11, 1154. [Google Scholar] [CrossRef] [PubMed]
- Buitimea-Cantúa, G.V.; Buitimea-Cantúa, N.E.; Rocha-Pizaña, M.d.R.; Hernández-Morales, A.; Magaña-Barajas, E.; Molina-Torres, J. Inhibitory effect of Capsicum chinense and Piper nigrum fruits, capsaicin and piperine on aflatoxins production in Aspergillus parasiticus by downregulating the expression of afl D, afl M, afl R, and afl S genes of aflatoxins biosynthetic pathway. J. Environ. Sci. Health Part B 2020, 55, 835–843. [Google Scholar] [CrossRef] [PubMed]
- Buitimea-Cantúa, G.V.; Velez-Haro, J.M.; Buitimea-Cantúa, N.E.; Molina-Torres, J.; Rosas-Burgos, E.C. GC-EIMS analysis, antifungal and anti-aflatoxigenic activity of Capsicum chinense and Piper nigrum fruits and their bioactive compounds capsaicin and piperine upon Aspergillus parasiticus. Nat. Prod. Res. 2020, 34, 1452–1455. [Google Scholar] [CrossRef]
- Millsop, J.W.; Fazel, N. Oral candidiasis. Clin. Dermatol. 2016, 34, 487–494. [Google Scholar] [CrossRef]
- Hurley, R.; De Louvois, J. Candida vaginitis. Postgrad. Med. J. 1979, 55, 645–647. [Google Scholar] [CrossRef]
- Sobel, J.D. Vulvovaginal candidosis. Lancet 2007, 369, 1961–1971. [Google Scholar] [CrossRef]
- Mayer, F.L.; Wilson, D.; Hube, B. Candida albicans pathogenicity mechanisms. Virulence 2013, 4, 119–128. [Google Scholar] [CrossRef]
- Fidel, P.L., Jr. History and new insights into host defense against vaginal candidiasis. Trends Microbiol. 2004, 12, 220–227. [Google Scholar] [CrossRef]
- Pappas, P.G.; Kauffman, C.A.; Andes, D.; Benjamin, D.K., Jr.; Calandra, T.F.; Edwards, J.E., Jr.; Filler, S.G.; Fisher, J.F.; Kullberg, B.-J.; Zeichner, L.O.; et al. Clinical Practice Guidelines for the Management Candidiasis: 2009 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2009, 48, 503–535. [Google Scholar] [CrossRef] [PubMed]
- Pfaller, M.A.; Diekema, D.J. Epidemiology of invasive candidiasis: A persistent public health problem. Clin. Microbiol. Rev. 2007, 20, 133–163. [Google Scholar] [CrossRef] [PubMed]
- Pfaller, M.A.; Diekema, D.J. Epidemiology of invasive mycoses in North America. Crit. Rev. Microbiol. 2010, 36, 1–53. [Google Scholar] [CrossRef] [PubMed]
- Behbehani, J.M.; Irshad, M.; Shreaz, S.; Karched, M. Anticandidal Activity of Capsaicin and Its Effect on Ergosterol Biosynthesis and Membrane Integrity of Candida albicans. Int. J. Mol. Sci. 2023, 24, 1046. [Google Scholar] [CrossRef]
- Georgianna, D.R.; Payne, G.A. Genetic regulation of aflatoxin biosynthesis: From gene to genome. Fungal Genet. Biol. 2009, 46, 113–125. [Google Scholar] [CrossRef]
- Astoreca, A.; Vaamonde, G.; Dalcero, A.; Ramos, A.J.; Marín, S. Modelling the effect of temperature and water activity of Aspergillus flavus isolates from corn. Int. J. Food Microbiol. 2012, 156, 60–67. [Google Scholar] [CrossRef]
- Nikolić, M.; Savić, I.; Nikolić, A.; Jauković, M.; Kandić, V.; Stevanović, M.; Stanković, S. Toxigenic species Aspergillus parasiticus originating from Maize Kernels grown in Serbia. Toxins 2021, 13, 847. [Google Scholar] [CrossRef]
- Sebők, F.; Dobolyi, C.; Zágoni, D.; Risa, A.; Krifaton, C.; Hartman, M.; Cserháti, M.; Szoboszlay, S.; Kriszt, B. Aflatoxigenic Aspergillus flavus and Aspergillus parasiticus strains in Hungarian maize fields. Acta Microbiol. Immunol. Hung. 2016, 63, 491–502. [Google Scholar] [CrossRef]
- Hernández-Téllez, C.N.; Luque-Alcaraz, A.G.; Núñez-Mexía, S.A.; Cortez-Rocha, M.O.; Lizardi-Mendoza, J.; Rosas-Burgos, E.C.; Rosas-Durazo, A.J.; Parra-Vergara, N.V.; Plascencia-Jatomea, M. Relationship between the Antifungal Activity of Chitosan-Capsaicin Nanoparticles and the Oxidative Stress Response on Aspergillus parasiticus. Polymers 2022, 14, 2774. [Google Scholar] [CrossRef]
- Milder, J.E.; Walzer, P.D.; Kilgore, G.; Rutherford, I.; Klein, M. Clinical features of Strongyloides stercoralis infection in an endemic area of the United States. Gastroenterology 1981, 80, 1481–1488. [Google Scholar] [CrossRef]
- Marsh, K.; Snow, R.W. Host—Parasite interaction and morbidity in malaria endemic areas. Philos. Trans. R. Soc. London. Ser. B Biol. Sci. 1997, 352, 1385–1394. [Google Scholar] [CrossRef]
- Barry, M.A.; Weatherhead, J.E.; Hotez, P.J.; Woc-Colburn, L. Childhood parasitic infections endemic to the United States. Pediatr. Clin. 2013, 60, 471–485. [Google Scholar] [CrossRef]
- Mata, L. Sociocultural Factors in the Control and Prevention of Parasitic Diseases. Rev. Infect. Dis. 1982, 4, 871–879. [Google Scholar] [CrossRef] [PubMed]
- Fèvre, E.M.; Bronsvoort, B.M.d.C.; Hamilton, K.A.; Cleaveland, S. Animal movements and the spread of infectious diseases. Trends Microbiol. 2006, 14, 125–131. [Google Scholar] [CrossRef]
- Torgerson, P.R. One world health: Socioeconomic burden and parasitic disease control priorities. Vet. Parasitol. 2013, 195, 223–232. [Google Scholar] [CrossRef]
- Torgerson, P.R.; de Silva, N.R.; Fèvre, E.M.; Kasuga, F.; Rokni, M.B.; Zhou, X.-N.; Sripa, B.; Gargouri, N.; Willingham, A.L.; Stein, C. The global burden of foodborne parasitic diseases: An update. Trends Parasitol. 2014, 30, 20–26. [Google Scholar] [CrossRef] [PubMed]
- Pisarski, K. The global burden of disease of zoonotic parasitic diseases: Top 5 contenders for priority consideration. Trop. Med. Infect. Dis. 2019, 4, 44. [Google Scholar] [CrossRef]
- Sangster, N.; Batterham, P.; Chapman, H.D.; Duraisingh, M.; Le Jambre, L.; Shirley, M.; Upcroft, J.; Upcroft, P. Resistance to antiparasitic drugs: The role of molecular diagnosis. Int. J. Parasitol. 2002, 32, 637–653. [Google Scholar] [CrossRef]
- Geary, T.G.; Thompson, D.P. Development of antiparasitic drugs in the 21st century. Vet. Parasitol. 2003, 115, 167–184. [Google Scholar] [CrossRef] [PubMed]
- Periferakis, A.; Caruntu, A.; Periferakis, A.-T.; Scheau, A.-E.; Badarau, I.A.; Caruntu, C.; Scheau, C. Availability, Toxicology and Medical Significance of Antimony. Int. J. Environ. Res. Public Health 2022, 19, 4669. [Google Scholar] [CrossRef] [PubMed]
- Valera-Vera, E.A.; Reigada, C.; Sayé, M.; Digirolamo, F.A.; Galceran, F.; Miranda, M.R.; Pereira, C.A. Effect of capsaicin on the protozoan parasite Trypanosoma cruzi. FEMS Microbiol. Lett. 2020, 367, fnaa194. [Google Scholar] [CrossRef]
- Lourido, S. Toxoplasma gondii. Trends Parasitol. 2019, 35, 944–945. [Google Scholar] [CrossRef]
- Zhang, Y.; Lai, B.S.; Juhas, M.; Zhang, Y. Toxoplasma gondii secretory proteins and their role in invasion and pathogenesis. Microbiol. Res. 2019, 227, 126293. [Google Scholar] [CrossRef] [PubMed]
- Lima, T.S.; Lodoen, M.B. Mechanisms of human innate immune evasion by Toxoplasma gondii. Front. Cell. Infect. Microbiol. 2019, 9, 103. [Google Scholar] [CrossRef] [PubMed]
- Dubey, J.P.; Lindsay, D.S.; Speer, C.A. Structures of Toxoplasma gondii tachyzoites, bradyzoites, and sporozoites and biology and development of tissue cysts. Clin. Microbiol. Rev. 1998, 11, 267–299. [Google Scholar] [CrossRef]
- Konstantinovic, N.; Guegan, H.; Stäjner, T.; Belaz, S.; Robert-Gangneux, F. Treatment of toxoplasmosis: Current options and future perspectives. Food Waterborne Parasitol. 2019, 15, e00036. [Google Scholar] [CrossRef]
- Dardé, M.L.; Villena, I.; Pinon, J.M.; Beguinot, I. Severe toxoplasmosis caused by a Toxoplasma gondii strain with a new isoenzyme type acquired in French Guyana. J. Clin. Microbiol. 1998, 36, 324. [Google Scholar] [CrossRef]
- Silva, L.A.; Reis-Cunha, J.L.; Bartholomeu, D.C.; Vítor, R.W. Genetic Polymorphisms and Phenotypic Profiles of Sulfadiazine-Resistant and Sensitive Toxoplasma gondii Isolates Obtained from Newborns with Congenital Toxoplasmosis in Minas Gerais, Brazil. PLoS ONE 2017, 12, e0170689. [Google Scholar] [CrossRef] [PubMed]
- Simarro, P.P.; Cecchi, G.; Paone, M.; Franco, J.R.; Diarra, A.; Ruiz, J.A.; Fèvre, E.M.; Courtin, F.; Mattioli, R.C.; Jannin, J.G. The Atlas of human African trypanosomiasis: A contribution to global mapping of neglected tropical diseases. Int. J. Health Geogr. 2010, 9, 57. [Google Scholar] [CrossRef]
- Desquesnes, M.; Dia, M.L. Trypanosoma vivax: Mechanical transmission in cattle by one of the most common African tabanids, Atylotus agrestis. Exp. Parasitol. 2003, 103, 35–43. [Google Scholar] [CrossRef]
- Desquesnes, M.; Dia, M.L. Mechanical transmission of Trypanosoma congolense in cattle by the African tabanid Atylotus agrestis. Exp. Parasitol. 2003, 105, 226–231. [Google Scholar] [CrossRef] [PubMed]
- Bouteille, B.; Oukem, O.; Bisser, S.; Dumas, M. Treatment perspectives for human African trypanosomiasis. Fundam. Clin. Pharmacol. 2003, 17, 171–181. [Google Scholar] [CrossRef] [PubMed]
- Baker, N.; de Koning, H.P.; Mäser, P.; Horn, D. Drug resistance in African trypanosomiasis: The melarsoprol and pentamidine story. Trends Parasitol. 2013, 29, 110–118. [Google Scholar] [CrossRef]
- Lavanchy, D. Hepatitis B virus epidemiology, disease burden, treatment, and current and emerging prevention and control measures. J. Viral Hepat. 2004, 11, 97–107. [Google Scholar] [CrossRef]
- Ly, K.N.; Xing, J.; Klevens, R.M.; Jiles, R.B.; Ward, J.W.; Holmberg, S.D. The increasing burden of mortality from viral hepatitis in the United States between 1999 and 2007. Ann. Intern. Med. 2012, 156, 271–278. [Google Scholar] [CrossRef] [PubMed]
- Kirk, M.D.; Pires, S.M.; Black, R.E.; Caipo, M.; Crump, J.A.; Devleesschauwer, B.; Döpfer, D.; Fazil, A.; Fischer-Walker, C.L.; Hald, T. World Health Organization estimates of the global and regional disease burden of 22 foodborne bacterial, protozoal, and viral diseases, 2010: A data synthesis. PLoS Med. 2015, 12, e1001921. [Google Scholar]
- Savic, M.; Penders, Y.; Shi, T.; Branche, A.; Pirçon, J.Y. Respiratory syncytial virus disease burden in adults aged 60 years and older in high-income countries: A systematic literature review and meta-analysis. Influenza Other Respir. Viruses 2023, 17, e13031. [Google Scholar] [CrossRef]
- Manolescu, L.; Marinescu, P.; Sultana, C.; Temereanca, A.; Vagu, C.; Grancea, C.; Ruta, S. HIV lamivudine resistance mutations in HBV co-infected Romanian patients. J. Int. AIDS Soc. 2008, 11, P182. [Google Scholar] [CrossRef]
- Temereanca, A.; Ene, L.; Sultana, C.; Manolescu, L.; Duiculescu, D.; Ruta, S. Transmitted HIV-1 drug resistance in untreated Romanian patients. In Proceedings of the Infectious Diseases Society of America 2011 Annual Meeting, Boston, MA, USA, 20–23 October 2011. [Google Scholar]
- Sultana, C.; Oprisan, G.; Szmal, C.; Vagu, C.; Temereanca, A.; Dinu, S.; Teleman, M.D.; Ruta, S. Molecular epidemiology of hepatitis C virus strains from Romania. J. Gastrointestin. Liver Dis. 2011, 20, 261–266. [Google Scholar]
- Lefeuvre, P.; Moriones, E. Recombination as a motor of host switches and virus emergence: Geminiviruses as case studies. Curr. Opin. Virol. 2015, 10, 14–19. [Google Scholar] [CrossRef]
- Bo, Z.; Miao, Y.; Xi, R.; Gao, X.; Miao, D.; Chen, H.; Jung, Y.S.; Qian, Y.; Dai, J. Emergence of a novel pathogenic recombinant virus from Bartha vaccine and variant pseudorabies virus in China. Transbound. Emerg. Dis. 2021, 68, 1454–1464. [Google Scholar] [CrossRef]
- Sultana, C.; Casian, M.; Oprea, C.; Ianache, I.; Grancea, C.; Chiriac, D.; Ruta, S. Hepatitis B Virus Genotypes and Antiviral Resistance Mutations in Romanian HIV-HBV Co-Infected Patients. Medicina 2022, 58, 531. [Google Scholar] [CrossRef] [PubMed]
- Grossarth-Maticek, R.; Frentzel-Beyme, R.; Kanazir, D.; Jankovic, M.; Vetter, H. Reported herpes-virus-infection, fever and cancer incidence in a prospective study. J. Chronic Dis. 1987, 40, 967–976. [Google Scholar] [CrossRef] [PubMed]
- Parkin, D.M.; Bray, F. The burden of HPV-related cancers. Vaccine 2006, 24, S11–S25. [Google Scholar] [CrossRef] [PubMed]
- Sultana, C.; Oprişan, G.; Teleman, M.D.; Dinu, S.; Oprea, C.; Voiculescu, M.; Ruta, S.; Team, H.P. Impact of hepatitis C virus core mutations on the response to interferon-based treatment in chronic hepatitis C. World J. Gastroenterol. 2016, 22, 8406. [Google Scholar] [CrossRef] [PubMed]
- Goncalves, P.H.; Ziegelbauer, J.; Uldrick, T.S.; Yarchoan, R. Kaposi-sarcoma herpesvirus associated cancers and related diseases. Curr. Opin. HIV AIDS 2017, 12, 47. [Google Scholar] [CrossRef] [PubMed]
- Sultana, C.; Rosca, A.; Ruta, S. Correlation Between miR-125b Expression and Liver Fibrosis in Patients with Chronic Hepatitis C. Hepat. Mon. 2019, 19, e84615. [Google Scholar] [CrossRef]
- Szymonowicz, K.A.; Chen, J. Biological and clinical aspects of HPV-related cancers. Cancer Biol. Med. 2020, 17, 864. [Google Scholar] [CrossRef] [PubMed]
- Pavel, B.; Moroti, R.; Spataru, A.; Popescu, M.R.; Panaitescu, A.M.; Zagrean, A.-M. Neurological Manifestations of SARS-CoV2 Infection: A Narrative Review. Brain Sci. 2022, 12, 1531. [Google Scholar] [CrossRef]
- Fertig, T.E.; Chitoiu, L.; Terinte-Balcan, G.; Peteu, V.-E.; Marta, D.; Gherghiceanu, M. The atomic portrait of SARS-CoV-2 as captured by cryo-electron microscopy. J. Cell. Mol. Med. 2022, 26, 25–34. [Google Scholar] [CrossRef]
- Brauburger, K.; Hume, A.J.; Mühlberger, E.; Olejnik, J. Forty-five years of Marburg virus research. Viruses 2012, 4, 1878–1927. [Google Scholar] [CrossRef]
- Messaoudi, I.; Amarasinghe, G.K.; Basler, C.F. Filovirus pathogenesis and immune evasion: Insights from Ebola virus and Marburg virus. Nat. Rev. Microbiol. 2015, 13, 663–676. [Google Scholar] [CrossRef] [PubMed]
- Malvy, D.; McElroy, A.K.; de Clerck, H.; Günther, S.; van Griensven, J. Ebola virus disease. Lancet 2019, 393, 936–948. [Google Scholar] [CrossRef] [PubMed]
- Jacob, S.T.; Crozier, I.; Fischer, W.A.; Hewlett, A.; Kraft, C.S.; Vega, M.-A.d.L.; Soka, M.J.; Wahl, V.; Griffiths, A.; Bollinger, L. Ebola virus disease. Nat. Rev. Dis. Primers 2020, 6, 13. [Google Scholar] [CrossRef] [PubMed]
- Brunker, K.; Mollentze, N. Rabies virus. Trends Microbiol. 2018, 26, 886–887. [Google Scholar] [CrossRef] [PubMed]
- Wunner, W.H.; Conzelmann, K.-K. Rabies virus. In Rabies; Elsevier: Amsterdam, The Netherlands, 2020; pp. 43–81. [Google Scholar]
- Sedighpour, D.; Taghizadeh, H. The effects of mutation on the drug binding affinity of Neuraminidase: Case study of Capsaicin using steered molecular dynamics simulation. J. Mol. Model. 2022, 28, 36. [Google Scholar] [CrossRef] [PubMed]
- Tang, K.; Zhang, X.; Guo, Y. Identification of the dietary supplement capsaicin as an inhibitor of Lassa virus entry. Acta. Pharm. Sin. B 2020, 10, 789–798. [Google Scholar] [CrossRef]
- Labella, A.M.; Merel, S.E. Influenza. Med. Clin. N. Am. 2013, 97, 621–645. [Google Scholar] [CrossRef]
- Webster, R.G.; Govorkova, E.A. Continuing challenges in influenza. Ann. N. Y. Acad. Sci. 2014, 1323, 115–139. [Google Scholar] [CrossRef]
- Gaitonde, D.Y.; Moore, F.C.; Morgan, M.K. Influenza: Diagnosis and treatment. Am. Fam. Physician 2019, 100, 751–758. [Google Scholar]
- Wu, N.C.; Wilson, I.A. Influenza Hemagglutinin Structures and Antibody Recognition. Cold Spring Harb. Perspect. Med. 2020, 10. [Google Scholar] [CrossRef]
- Chang, C.; Ramphul, K. Amantadine. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2023. [Google Scholar]
- Palomba, E.; Castelli, V.; Renisi, G.; Bandera, A.; Lombardi, A.; Gori, A. Antiviral Treatments for Influenza. Semin Respir. Crit. Care Med. 2021, 42, 859–872. [Google Scholar] [CrossRef] [PubMed]
- Świerczyńska, M.; Mirowska-Guzel, D.M.; Pindelska, E. Antiviral Drugs in Influenza. Int. J. Environ. Res. Public Health 2022, 19, 3018. [Google Scholar] [CrossRef]
- Aschenbrenner, D.S. Xofluza Now Indicated to Prevent Influenza. Am. J. Nurs. 2021, 121, 26–27. [Google Scholar] [CrossRef]
- Tanret, I.; Duh, D. The Reye syndrome. J. Pharm. Belg. 2011, 1, 13–15. [Google Scholar]
- Chang, A.Y.; Mann, T.S.; McFawn, P.K.; Han, L.; Dong, X.; Henry, P.J. Investigating the role of MRGPRC11 and capsaicin-sensitive afferent nerves in the anti-influenza effects exerted by SLIGRL-amide in murine airways. Respir. Res. 2016, 17, 62. [Google Scholar] [CrossRef]
- Günther, S.; Lenz, O. Lassa virus. Crit. Rev. Clin. Lab. Sci. 2004, 41, 339–390. [Google Scholar] [CrossRef] [PubMed]
- Happi, A.N.; Happi, C.T.; Schoepp, R.J. Lassa fever diagnostics: Past, present, and future. Curr. Opin. Virol. 2019, 37, 132–138. [Google Scholar] [CrossRef]
- Lukashevich, I.S.; Paessler, S.; de la Torre, J.C. Lassa virus diversity and feasibility for universal prophylactic vaccine. F1000Res 2019, 8. [Google Scholar] [CrossRef]
- Alcorn, J.B. Huastec Mayan Ethnobotany; University of Texas Press: Austin, TX, USA, 1984. [Google Scholar]
- Jain, S. Ethnobotany. Interdiscip. Sci. Rev. 1986, 11, 285–292. [Google Scholar] [CrossRef]
- Cotton, C.M. Ethnobotany: Principles and Applications; John Wiley & Sons: Hoboken, NJ, USA, 1996. [Google Scholar]
- Heinrich, M. Ethnobotany and its role in drug development. Phytother. Res. Int. J. Devoted Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 2000, 14, 479–488. [Google Scholar] [CrossRef]
- Petran, M.; Dragos, D.; Gilca, M. Historical ethnobotanical review of medicinal plants used to treat children diseases in Romania (1860s–1970s). J. Ethnobiol. Ethnomedicine 2020, 16, 15. [Google Scholar] [CrossRef] [PubMed]
- Balick, M.J.; Cox, P.A. Plants, People, and Culture: The Science of Ethnobotany; Garland Science: New York, NY, USA, 2020. [Google Scholar]
- Teow, S.-Y.; Ali, S.A. Synergistic antibacterial activity of Curcumin with antibiotics against Staphylococcus aureus. Pak. J. Pharm. Sci. 2015, 28, 2109–2114. [Google Scholar]
- Yun, D.G.; Lee, D.G. Antibacterial activity of curcumin via apoptosis-like response in Escherichia coli. Appl. Microbiol. Biotechnol. 2016, 100, 5505–5514. [Google Scholar] [CrossRef] [PubMed]
- Gunes, H.; Gulen, D.; Mutlu, R.; Gumus, A.; Tas, T.; Topkaya, A.E. Antibacterial effects of curcumin: An in vitro minimum inhibitory concentration study. Toxicol. Ind. Health 2016, 32, 246–250. [Google Scholar] [CrossRef]
- Liao, Y.; Yao, Y.; Yu, Y.; Zeng, Y. Enhanced antibacterial activity of curcumin by combination with metal ions. Colloid Interface Sci. Commun. 2018, 25, 1–6. [Google Scholar] [CrossRef]
- Smyth, T.; Ramachandran, V.N.; Smyth, W.F. A study of the antimicrobial activity of selected naturally occurring and synthetic coumarins. Int. J. Antimicrob. Agents 2009, 33, 421–426. [Google Scholar] [CrossRef]
- Choo, S.; Chin, V.K.; Wong, E.H.; Madhavan, P.; Tay, S.T.; Yong, P.V.C.; Chong, P.P. Review: Antimicrobial properties of allicin used alone or in combination with other medications. Folia Microbiol. 2020, 65, 451–465. [Google Scholar] [CrossRef]
- Lauritano, D.; Petruzzi, M.; Baldoni, M. Preliminary protocol for systemic administration of capsaicin for the treatment of the burning mouth syndrome. Minerva Stomatol. 2003, 52, 273–278. [Google Scholar]
- Laude-Pagniez, E.; Leclerc, J.; Lok, C.; Chaby, G.; Arnault, J.P. Capsaicin 8% patch as therapy for neuropathic chronic postsurgical pain after melanoma excision surgery: A single center case series. JAAD Case Rep. 2022, 30, 70–75. [Google Scholar] [CrossRef]
- Yang, S.; Liu, L.; Meng, L.; Hu, X. Capsaicin is beneficial to hyperlipidemia, oxidative stress, endothelial dysfunction, and atherosclerosis in Guinea pigs fed on a high-fat diet. Chem. Biol. Interact. 2019, 297, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Chan, T.C.; Lee, M.S.; Huang, W.C.; Chang, W.Y.; Krueger, J.G.; Tsai, T.F. Capsaicin attenuates imiquimod-induced epidermal hyperplasia and cutaneous inflammation in a murine model of psoriasis. Biomed. Pharmacother. 2021, 141, 111950. [Google Scholar] [CrossRef] [PubMed]
- Thán, M.; Németh, J.; Szilvássy, Z.; Pintér, E.; Helyes, Z.; Szolcsányi, J. Systemic anti-inflammatory effect of somatostatin released from capsaicin-sensitive vagal and sciatic sensory fibres of the rat and guinea-pig. Eur. J. Pharmacol. 2000, 399, 251–258. [Google Scholar] [CrossRef] [PubMed]
- Popescu, G.D.A.; Scheau, C.; Badarau, I.A.; Dumitrache, M.D.; Caruntu, A.; Scheau, A.E.; Costache, D.O.; Costache, R.S.; Constantin, C.; Neagu, M.; et al. The Effects of Capsaicin on Gastrointestinal Cancers. Molecules 2020, 26, 94. [Google Scholar] [CrossRef]
- Han, T.H.; Park, M.K.; Nakamura, H.; Ban, H.S. Capsaicin inhibits HIF-1α accumulation through suppression of mitochondrial respiration in lung cancer cells. Biomed. Pharmacother. 2022, 146, 112500. [Google Scholar] [CrossRef]
- Liu, X.-Y.; Wei, D.-G.; Li, R.-S. Capsaicin induces ferroptosis of NSCLC by regulating SLC7A11/GPX4 signaling in vitro. Sci. Rep. 2022, 12, 11996. [Google Scholar] [CrossRef]
- Que, T.; Ren, B.; Fan, Y.; Liu, T.; Hou, T.; Dan, W.; Liu, B.; Wei, Y.; Lei, Y.; Zeng, J.; et al. Capsaicin inhibits the migration, invasion and EMT of renal cancer cells by inducing AMPK/mTOR-mediated autophagy. Chem. Biol. Interact. 2022, 366, 110043. [Google Scholar] [CrossRef]
- Scheau, C.; Badarau, I.A.; Caruntu, C.; Mihai, G.L.; Didilescu, A.C.; Constantin, C.; Neagu, M. Capsaicin: Effects on the Pathogenesis of Hepatocellular Carcinoma. Molecules 2019, 24, 2350. [Google Scholar] [CrossRef]
- Georgescu, S.-R.; Sârbu, M.-I.; Matei, C.; Ilie, M.A.; Caruntu, C.; Constantin, C.; Neagu, M.; Tampa, M. Capsaicin: Friend or Foe in Skin Cancer and Other Related Malignancies? Nutrients 2017, 9, 1365. [Google Scholar] [CrossRef]
- Scheau, C.; Mihai, L.; Bădărău, I.; Caruntu, C. Emerging applications of some important natural compounds in the field of oncology. Farmacia 2020, 68, 984–991. [Google Scholar] [CrossRef]
- Redington, K.L.; Disenhouse, T.; Strantzas, S.C.; Gladstone, R.; Wei, C.; Tropak, M.B.; Dai, X.; Manlhiot, C.; Li, J.; Redington, A.N. Remote cardioprotection by direct peripheral nerve stimulation and topical capsaicin is mediated by circulating humoral factors. Basic Res. Cardiol. 2012, 107, 241. [Google Scholar] [CrossRef]
- Scheau, C.; Ilie Ghita, M.; Grigore, O.; Mihailescu, A.; Caruntu, A.; Mihai, L.; Bădărău, I.; Boda, D.; Caruntu, C. Modulation of capsaicin-induced neurogenic vasodilation by acute psychological stress. Farmacia 2021, 69, 778–784. [Google Scholar] [CrossRef]
- Shin, M.K.; Yang, S.-M.; Han, I.-S. Capsaicin suppresses liver fat accumulation in high-fat diet-induced NAFLD mice. Anim. Cells Syst. 2020, 24, 214–219. [Google Scholar] [CrossRef] [PubMed]
- Yu, Q.; Wang, Y.; Yu, Y.; Li, Y.; Zhao, S.; Chen, Y.; Waqar, A.B.; Fan, J.; Liu, E. Expression of TRPV1 in rabbits and consuming hot pepper affects its body weight. Mol. Biol. Rep. 2012, 39, 7583–7589. [Google Scholar] [CrossRef]
- Dumitrache, M.D.; Jieanu, A.S.; Scheau, C.; Badarau, I.A.; Popescu, G.D.A.; Caruntu, A.; Costache, D.O.; Costache, R.S.; Constantin, C.; Neagu, M.; et al. Comparative effects of capsaicin in chronic obstructive pulmonary disease and asthma (Review). Exp. Ther. Med. 2021, 22, 917. [Google Scholar] [CrossRef]
- Aguilar-Meléndez, A.; Vásquez, M.A.; Katz, E.; Colorado, M.R.H. Los Chiles Que le Dan Sabor al Mundo: Contribuciones Multidisciplinarias; IRD Éditions: Hong Kong, China, 2018.
- Aguilar-Meléndez, A.; Vásquez-Dávila, M.A.; Manzanero-Medina, G.I.; Katz, E. Chile (Capsicum spp.) as Food-Medicine Continuum in Multiethnic Mexico. Foods 2021, 10, 2502. [Google Scholar] [CrossRef] [PubMed]
- Sarwa, K. Chapter 04—Some Indigenous Capsicum Species of India And Their Ethnomedicinal Importance Government of India. In Recent Advances in Ethnobotany; Deep Publication: New Delhi, India, 2021; pp. 18–21. [Google Scholar]
- Saleh, B.K.; Omer, A.; Teweldemedhin, B. Medicinal uses and health benefits of chili pepper (Capsicum spp.): A review. MOJ Food Process Technol. 2018, 6, 325–328. [Google Scholar] [CrossRef]
- Meghvansi, M.K.; Siddiqui, S.; Khan, M.H.; Gupta, V.K.; Vairale, M.G.; Gogoi, H.K.; Singh, L. Naga chilli: A potential source of capsaicinoids with broad-spectrum ethnopharmacological applications. J. Ethnopharmacol. 2010, 132, 1–14. [Google Scholar] [CrossRef]
- Sayanlar, J.; Guleyupoglu, N.; Portenoy, R.; Ashina, S. Trigeminal postherpetic neuralgia responsive to treatment with capsaicin 8% topical patch: A case report. J. Headache. Pain 2012, 13, 587–589. [Google Scholar] [CrossRef]
- Zis, P.; Apsokardos, A.; Isaia, C.; Sykioti, P.; Vadalouca, A. Posttraumatic and postsurgical neuropathic pain responsive to treatment with capsaicin 8% topical patch. Pain. Physician 2014, 17, E213–E218. [Google Scholar]
- Zeidler, C.; Metze, D.; Ständer, S. Successful treatment of lichen amyloidosis using capsaicin 8% patch. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 1236–1238. [Google Scholar] [CrossRef] [PubMed]
- Kocak, A.O.; Dogruyol, S.; Akbas, I.; Menekse, T.S.; Gur, S.T.A.; Kocak, M.B.; Cekmen, B.; Orun, S.; Cakir, Z. Comparison of topical capsaicin and topical piroxicam in the treatment of acute trauma-induced pain: A randomized double-blind trial. Am. J.. Emerg. Med. 2020, 38, 1767–1771. [Google Scholar] [CrossRef] [PubMed]
- Hoesli, R.C.; Wingo, M.L.; Wajsberg, B.; Bastian, R.W. Topical Capsaicin for the Treatment of Sensory Neuropathic Cough. OTO Open 2021, 5, 2473974x211065668. [Google Scholar] [CrossRef] [PubMed]
- Van Gerven, L.; Steelant, B.; Cools, L.; Callebaut, I.; Backaert, W.; de Hoon, J.; Ampe, E.; Talavera, K.; Hellings, P.W. Low-dose capsaicin (0.01 mM) nasal spray is equally effective as the current standard treatment for idiopathic rhinitis: A randomized, double-blind, placebo-controlled trial. J. Allergy Clin. Immunol. 2021, 147, 397–400.e394. [Google Scholar] [CrossRef]
- Vachiramon, V.; Tanratana, P.; Anuntrangsee, T.; Palakornkitti, P.; Yeesibsean, N.; Kungvalpivat, P.; Fabi, S. The role of topical capsaicin gel in pain management during microfocused ultrasound treatment for neck laxity. Ski. Res. Technol. 2023, 29, e13240. [Google Scholar] [CrossRef]
- Hägermark, O.; Hökfelt, T.; Pernow, B. Flare and itch induced by substance P in human skin. Nation 1978, 12, 13. [Google Scholar] [CrossRef] [PubMed]
- Bartold, P.; Kylstra, A.; Lawson, R. Substance P: An immunohistochemical and biochemical study in human gingival tissues. A role for neurogenic inflammation? J. Periodontol. 1994, 65, 1113–1121. [Google Scholar] [CrossRef]
- Figini, M.; Emanueli, C.; Bertrand, C.; Javdan, P.; Geppetti, P. Evidence that tachykinins relax the guinea-pig trachea via nitric oxide release and by stimulation of a septide-insensitive NK1 receptor. Br. J. Pharmacol. 1996, 117, 1270–1276. [Google Scholar] [CrossRef]
- Simone, D.A.; Nolano, M.; Johnson, T.; Wendelschafer-Crabb, G.; Kennedy, W.R. Intradermal injection of capsaicin in humans produces degeneration and subsequent reinnervation of epidermal nerve fibers: Correlation with sensory function. J. Neurosci. 1998, 18, 8947–8959. [Google Scholar] [CrossRef]
- Chen, F.X.; Wan, Q.; Fang, J.; Peng, L.; Li, Q.L.; Hu, J. The Src1-PGC1α-AP1 complex-dependent secretion of substance P induces inflammation and apoptosis in encephalomyocarditis virus-infected mice. Cytokine 2023, 165, 156186. [Google Scholar] [CrossRef]
- Johnson, M.B.; Suptela, S.R.; Sipprell, S.E.; Marriott, I. Substance P Exacerbates the Inflammatory and Pro-osteoclastogenic Responses of Murine Osteoclasts and Osteoblasts to Staphylococcus aureus. Inflammation 2023, 46, 256–269. [Google Scholar] [CrossRef] [PubMed]
- Arruda-Vasconcelos, R.; Chiarelli-Neto, V.M.; Louzada, L.M.; Aveiro, E.; Alves-Silva, E.G.; de-Jesus-Soares, A.; Ferraz, C.C.R.; Almeida, J.F.A.; Marciano, M.A.; Pecorari, V.G.A.; et al. Quantitative analysis of culturable bacteria, levels of endotoxins, inflammatory mediators and substance P in teeth with symptomatic irreversible pulpitis and in teeth with vital normal pulp tissues. Int. Endod J. 2023, 56, 827–836. [Google Scholar] [CrossRef]
- Mehboob, R.; Oehme, P.; Pfaff, G. The role of Substance P in the defense line of the respiratory tract and neurological manifestations post COVID-19 infection. Front. Neurol. 2023, 14, 1052811. [Google Scholar] [CrossRef] [PubMed]
- Szolcsányi, J.; Barthó, L. Capsaicin-sensitive afferents and their role in gastroprotection: An update. J. Physiol. Paris 2001, 95, 181–188. [Google Scholar] [CrossRef]
- Szolcsányi, J.; Helyes, Z.; Oroszi, G.; Németh, J.; Pintér, E. Release of somatostatin and its role in the mediation of the anti-inflammatory effect induced by antidromic stimulation of sensory fibres of rat sciatic nerve. Br. J. Pharmacol. 1998, 123, 936–942. [Google Scholar] [CrossRef]
- Helyes, Z.; Thán, M.; Oroszi, G.; Pintér, E.; Németh, J.; Kéri, G.; Szolcsányi, J. Anti-nociceptive effect induced by somatostatin released from sensory nerve terminals and by synthetic somatostatin analogues in the rat. Neurosci. Lett. 2000, 278, 185–188. [Google Scholar] [CrossRef] [PubMed]
- Pintér, E.; Helyes, Z.; Szolcsányi, J. Inhibitory effect of somatostatin on inflammation and nociception. Pharmacol. Ther. 2006, 112, 440–456. [Google Scholar] [CrossRef]
- László, S.; Bátai, I.Z.; Berkó, S.; Csányi, E.; Dombi, Á.; Pozsgai, G.; Bölcskei, K.; Botz, L.; Wagner, Ö.; Pintér, E. Development of Capsaicin-Containing Analgesic Silicone-Based Transdermal Patches. Pharmaceuticals 2022, 15, 1279. [Google Scholar] [CrossRef] [PubMed]
- Peck, R. Neuropeptides modulating macrophage function. Ann. N. Y. Acad. Sci. 1987, 496, 264–270. [Google Scholar] [CrossRef]
- Weinstock, J.V.; Elliott, D. The somatostatin immunoregulatory circuit present at sites of chronic inflammation. Eur. J. Endocrinol. 2000, 143, S15–S19. [Google Scholar] [CrossRef]
- Kao, J.Y.; Pierzchala, A.; Rathinavelu, S.; Zavros, Y.; Tessier, A.; Merchant, J.L. Somatostatin inhibits dendritic cell responsiveness to Helicobacter pylori. Regul. Pept. 2006, 134, 23–29. [Google Scholar] [CrossRef]
- Monaco-Shawver, L.; Schwartz, L.; Tuluc, F.; Guo, C.-J.; Lai, J.P.; Gunnam, S.M.; Kilpatrick, L.E.; Banerjee, P.P.; Douglas, S.D.; Orange, J.S. Substance P inhibits natural killer cell cytotoxicity through the neurokinin-1 receptor. J. Leukoc. Biol. 2010, 89, 113–125. [Google Scholar] [CrossRef] [PubMed]
- Froidevaux, S.; Eberle, A.N. Somatostatin analogs and radiopeptides in cancer therapy. Pept. Sci. 2002, 66, 161–183. [Google Scholar] [CrossRef]
- Klironomos, S.; Notas, G.; Sfakianaki, O.; Kiagiadaki, F.; Xidakis, C.; Kouroumalis, E. Octreotide modulates the effects on fibrosis of TNF-α, TGF-β and PDGF in activated rat hepatic stellate cells. Regul. Pept. 2014, 188, 5–12. [Google Scholar] [CrossRef]
- Ayiomamitis, G.D.; Notas, G.; Zaravinos, A.; Drygiannakis, I.; Georgiadou, M.; Sfakianaki, O.; Mastrodimou, N.; Thermos, K.; Kouroumalis, E. Effects of octreotide and insulin on colon cancer cellular proliferation and correlation with hTERT activity. Oncoscience 2014, 1, 457. [Google Scholar] [CrossRef] [PubMed]
- Kouroumalis, E.; Samonakis, D.; Notas, G. Somatostatin in hepatocellular carcinoma: Experimental and therapeutic implications. Hepatoma Res. 2018, 4, 34. [Google Scholar] [CrossRef]
- Periferakis, A.; Tsigas, G.; Periferakis, A.-T.; Badarau, I.A.; Scheau, A.-E.; Tampa, M.; Georgescu, S.R.; Didilescu, A.C.; Scheau, C.; Caruntu, C. Antitumoral and Anti-inflammatory Roles of Somatostatin and Its Analogs in Hepatocellular Carcinoma. Anal. Cell. Pathol. 2021, 2021, 1840069. [Google Scholar] [CrossRef]
- Kouroumalis, E.; Tsomidis, I.; Voumvouraki, A. Is There a Place for Somatostatin Analogues for the Systemic Treatment of Hepatocellular Carcinoma in the Immunotherapy Era? Livers 2022, 2, 315–335. [Google Scholar] [CrossRef]
- McLaughlin-Drubin, M.E.; Munger, K. Viruses associated with human cancer. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2008, 1782, 127–150. [Google Scholar] [CrossRef] [PubMed]
- Georgakilas, A.G.; Mosley, W.G.; Georgakila, S.; Ziech, D.; Panayiotidis, M.I. Viral-induced human carcinogenesis: An oxidative stress perspective. Mol. BioSystems 2010, 6, 1162–1172. [Google Scholar] [CrossRef]
- Alibek, K.; Kakpenova, A.; Mussabekova, A.; Sypabekova, M.; Karatayeva, N. Role of viruses in the development of breast cancer. Infect. Agents Cancer 2013, 8, 32. [Google Scholar] [CrossRef]
- Hatano, Y.; Ideta, T.; Hirata, A.; Hatano, K.; Tomita, H.; Okada, H.; Shimizu, M.; Tanaka, T.; Hara, A. Virus-Driven Carcinogenesis. Cancers 2021, 13, 2625. [Google Scholar] [CrossRef]
- Brizzi, K.T.; Lyons, J.L. Peripheral nervous system manifestations of infectious diseases. Neurohospitalist 2014, 4, 230–240. [Google Scholar] [CrossRef]
- De León, A.M.; Garcia-Santibanez, R.; Harrison, T.B. Article Topic: Neuropathies Due to Infections and Antimicrobial Treatments. Curr. Treat Options Neurol. 2023, 1–17. [Google Scholar] [CrossRef]
- Peppin, J.F.; Pappagallo, M. Capsaicinoids in the treatment of neuropathic pain: A review. Ther. Adv. Neurol. Disord. 2014, 7, 22–32. [Google Scholar] [CrossRef]
- Filippi, A.; Caruntu, C.; Gheorghe, R.O.; Deftu, A.; Amuzescu, B.; Ristoiu, V. Catecholamines reduce transient receptor potential vanilloid type 1 desensitization in cultured dorsal root ganglia neurons. J. Physiol. Pharmacol. 2016, 67, 843–850. [Google Scholar] [PubMed]
- Mihai, A.; Chitimus, D.M.; Jurcut, C.; Blajut, F.C.; Opris-Belinski, D.; Caruntu, C.; Ionescu, R.; Caruntu, A. Comparative Analysis of Hematological and Immunological Parameters in Patients with Primary Sjögren’s Syndrome and Peripheral Neuropathy. J. Clin. Med. 2023, 12, 3672. [Google Scholar] [CrossRef] [PubMed]
- Kazamel, M.; Stino, A.M.; Smith, A.G. Metabolic syndrome and peripheral neuropathy. Muscle Nerve 2021, 63, 285–293. [Google Scholar] [CrossRef] [PubMed]
- Bucurica, S.; Prodan, I.; Pavalean, M.; Taubner, C.; Bucurica, A.; Socol, C.; Calin, R.; Ionita-Radu, F.; Jinga, M. Association of Vitamin D Deficiency and Insufficiency with Pathology in Hospitalized Patients. Diagnostics 2023, 13, 998. [Google Scholar] [CrossRef]
- Kawada, T.; Suzuki, T.; Takahashi, M.; Iwai, K. Gastrointestinal absorption and metabolism of capsaicin and dihydrocapsaicin in rats. Toxicol. Appl. Pharmacol. 1984, 72, 449–456. [Google Scholar] [CrossRef] [PubMed]
- Sharma, S.K.; Vij, A.S.; Sharma, M. Mechanisms and clinical uses of capsaicin. Eur. J. Pharmacol. 2013, 720, 55–62. [Google Scholar] [CrossRef] [PubMed]
- Chanda, S.; Bashir, M.; Babbar, S.; Koganti, A.; Bley, K. In vitro hepatic and skin metabolism of capsaicin. Drug Metab. Dispos. 2008, 36, 670–675. [Google Scholar] [CrossRef]
- Qin, L.; Wang, Y.; Gong, Y.; Chen, J.; Xu, B.; Tang, L.; Guo, L.; Xie, J. Capsaicin metabolites and GSH-associated detoxification and biotransformation pathways in human liver microsomes revealed by LC-HRMS/MS with data-mining tools. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2019, 1133, 121843. [Google Scholar] [CrossRef] [PubMed]
- Rollyson, W.D.; Stover, C.A.; Brown, K.C.; Perry, H.E.; Stevenson, C.D.; McNees, C.A.; Ball, J.G.; Valentovic, M.A.; Dasgupta, P. Bioavailability of capsaicin and its implications for drug delivery. J. Control. Release 2014, 196, 96–105. [Google Scholar] [CrossRef]
- Babbar, S.; Marier, J.F.; Mouksassi, M.S.; Beliveau, M.; Vanhove, G.F.; Chanda, S.; Bley, K. Pharmacokinetic analysis of capsaicin after topical administration of a high-concentration capsaicin patch to patients with peripheral neuropathic pain. Ther. Drug Monit. 2009, 31, 502–510. [Google Scholar] [CrossRef]
- Zak, A.; Siwinska, N.; Slowikowska, M.; Borowicz, H.; Szpot, P.; Zawadzki, M.; Niedzwiedz, A. The detection of capsaicin and dihydrocapsaicin in horse serum following long-term local administration. BMC Vet. Res. 2018, 14, 193. [Google Scholar] [CrossRef]
- Goci, E.; Haloci, E.; Di Stefano, A.; Chiavaroli, A.; Angelini, P.; Miha, A.; Cacciatore, I.; Marinelli, L. Evaluation of In Vitro Capsaicin Release and Antimicrobial Properties of Topical Pharmaceutical Formulation. Biomolecules 2021, 11, 432. [Google Scholar] [CrossRef] [PubMed]
- Reilly, C.A.; Yost, G.S. Metabolism of capsaicinoids by P450 enzymes: A review of recent findings on reaction mechanisms, bio-activation, and detoxification processes. Drug Metab. Rev. 2006, 38, 685–706. [Google Scholar] [CrossRef] [PubMed]
- van Eijl, S.; Zhu, Z.; Cupitt, J.; Gierula, M.; Götz, C.; Fritsche, E.; Edwards, R.J. Elucidation of xenobiotic metabolism pathways in human skin and human skin models by proteomic profiling. PLoS ONE 2012, 7, e41721. [Google Scholar] [CrossRef]
- Tian, K.; Zhu, J.; Li, M.; Qiu, X. Capsaicin is efficiently transformed by multiple cytochrome P450s from Capsicum fruit-feeding Helicoverpa armigera. Pestic. Biochem. Physiol. 2019, 156, 145–151. [Google Scholar] [CrossRef]
- Marquez-Algaba, E.; Burgos, J.; Almirante, B. Pharmacotherapeutic interventions for the treatment of bacterial prostatitis. Expert Opin. Pharmacother. 2022, 23, 1091–1101. [Google Scholar] [CrossRef]
- Norman, G.; Dumville, J.C.; Mohapatra, D.P.; Owens, G.L.; Crosbie, E.J. Antibiotics and antiseptics for surgical wounds healing by secondary intention. Cochrane Database Syst. Rev. 2016, 3, Cd011712. [Google Scholar] [CrossRef]
- Dragosloveanu, Ş.; Dragosloveanu, C.D.M.; Stanca, H.T.; Cotor, D.C.; Dragosloveanu, C.I.; Stoica, C.I. A new perspective towards failure of gamma nail systems. Exp. Ther. Med. 2020, 20, 216. [Google Scholar] [CrossRef] [PubMed]
- Monaghan, M.G.; Murphy, C.M. Old Drugs, New Tricks–Redefining Therapeutic Strategies for Tissue Regeneration. Adv. Drug Deliv. Rev. 2021, 173, 279–280. [Google Scholar] [CrossRef]
- Bergren, D.R. Capsaicin challenge, reflex bronchoconstriction, and local action of substance P. Am. J. Physiol. 1988, 254, R845–R852. [Google Scholar] [CrossRef] [PubMed]
- Thomas, K.C.; Ethirajan, M.; Shahrokh, K.; Sun, H.; Lee, J.; Cheatham, T.E., 3rd; Yost, G.S.; Reilly, C.A. Structure-activity relationship of capsaicin analogs and transient receptor potential vanilloid 1-mediated human lung epithelial cell toxicity. J. Pharmacol. Exp. Ther. 2011, 337, 400–410. [Google Scholar] [CrossRef]
- Trevisan, G.; Rossato, M.F.; Hoffmeister, C.; Oliveira, S.M.; Silva, C.R.; Matheus, F.C.; Mello, G.C.; Antunes, E.; Prediger, R.D.; Ferreira, J. Mechanisms involved in abdominal nociception induced by either TRPV1 or TRPA1 stimulation of rat peritoneum. Eur. J. Pharmacol. 2013, 714, 332–344. [Google Scholar] [CrossRef]
- Kwon, Y. Estimation of Dietary Capsaicinoid Exposure in Korea and Assessment of Its Health Effects. Nutrients 2021, 13, 2461. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Gu, Q.; Qu, C. Capsaicin pretreatment reversed pulmonary arterial hypertension by alleviating inflammation via p38MAPK pathway. Exp. Lung Res. 2017, 43, 8–18. [Google Scholar] [CrossRef]
- Lechner, A.; Alderson, T.; Gautam, S.; Flaker, G. Ventricular fibrillation due to coronary spasm after pepper spray. Pacing. Clin. Electrophysiol. 2021, 44, 548–551. [Google Scholar] [CrossRef] [PubMed]
- Jancsó, G.; Király, E.; Such, G.; Joó, F.; Nagy, A. Neurotoxic effect of capsaicin in mammals. Acta. Physiol. Hung. 1987, 69, 295–313. [Google Scholar] [PubMed]
- Ritter, S.; Dinh, T.T. Capsaicin-induced neuronal degeneration in the brain and retina of preweanling rats. J. Comp. Neurol. 1990, 296, 447–461. [Google Scholar] [CrossRef] [PubMed]
- Forrester, M.B.; Holloway, C. Characteristics of pepper spray-related injuries reported to the National Electronic Injury Surveillance System during 2000–2020. Clin. Toxicol. 2022, 60, 348–355. [Google Scholar] [CrossRef] [PubMed]
- McGilton, K.S.; Höbler, F.; Campos, J.; Dupuis, K.; Labreche, T.; Guthrie, D.M.; Jarry, J.; Singh, G.; Wittich, W. Hearing and vision screening tools for long-term care residents with dementia: Protocol for a scoping review. BMJ Open 2016, 6, e011945. [Google Scholar] [CrossRef] [PubMed]
- Stoica, C.I.; Nedelea, G.; Cotor, D.C.; Gherghe, M.; Georgescu, D.E.; Dragosloveanu, C.; Dragosloveanu, S. The Outcome of Total Knee Arthroplasty for Patients with Psychiatric Disorders: A Single-Center Retrospective Study. Medicina 2022, 58, 1277. [Google Scholar] [CrossRef]
- Sloane, P.D.; Whitson, H.; Williams, S.W. Addressing Hearing and Vision Impairment in Long-Term Care: An Important and Often-Neglected Care Priority. J. Am. Med. Dir. Assoc. 2021, 22, 1151–1155. [Google Scholar] [CrossRef]
- Ologunagba, M.; Kolawole, O.; Echerenwa, A.; Silva, B. Development and characterization of capsaicin creams formulated with Grewia mucilage-HPMC base. J. Sci. Pract. Pharm. 2023, 7, 365–375. [Google Scholar] [CrossRef]
- Trbojević Ivić, J.; Milosavić, N.; Dimitrijević, A.; Gavrović Jankulović, M.; Bezbradica, D.; Kolarski, D.; Veličković, D. Synthesis of medium-chain length capsinoids from coconut oil catalyzed by Candida rugosa lipases. Food Chem. 2017, 218, 505–508. [Google Scholar] [CrossRef]
- Tangwatcharin, P.; Khopaibool, P. Activity of virgin coconut oil, lauric acid or monolaurin in combination with lactic acid against Staphylococcus aureus. Southeast Asian J. Trop. Med. Public Health 2012, 43, 969–985. [Google Scholar]
- Shilling, M.; Matt, L.; Rubin, E.; Visitacion, M.P.; Haller, N.A.; Grey, S.F.; Woolverton, C.J. Antimicrobial effects of virgin coconut oil and its medium-chain fatty acids on Clostridium difficile. J. Med. Food 2013, 16, 1079–1085. [Google Scholar] [CrossRef]
- Peedikayil, F.C.; Remy, V.; John, S.; Chandru, T.P.; Sreenivasan, P.; Bijapur, G.A. Comparison of antibacterial efficacy of coconut oil and chlorhexidine on Streptococcus mutans: An in vivo study. J. Int. Soc. Prev. Community Dent. 2016, 6, 447–452. [Google Scholar] [CrossRef]
- Widianingrum, D.C.; Noviandi, C.T.; Salasia, S.I.O. Antibacterial and immunomodulator activities of virgin coconut oil (VCO) against Staphylococcus aureus. Heliyon 2019, 5, e02612. [Google Scholar] [CrossRef]
- Hariyadi, D.M.; Fitri, A.; Sudarma, S.; Purwanti, T.; Erawati, T. Optimization of microspheres containing virgin coconut oil and hydrolyzed virgin coconut oil as antimicrobial. J. Adv. Pharm. Technol. Res. 2022, 13, 238–242. [Google Scholar] [CrossRef] [PubMed]
- Elmore, L.K.; Nance, G.; Singleton, S.; Lorenz, L. Treatment of dermal infections with topical coconut oil. Nat. Med. J. 2014, 6. Available online: https://www.naturalmedicinejournal.com/journal/treatment-dermal-infections-topical-coconut-oil (accessed on 12 September 2023).
- Lassen, C.L.; Meyer, K.; Bredthauer, A.; Klier, T.W. Facial and Oral Cross-Contamination of a 3-Year-Old Child with High Concentration Capsaicin: A Case Report. A A Pract. 2020, 14, e01258. [Google Scholar] [CrossRef] [PubMed]
- Yeung, M.F.; Tang, W.Y. Clinicopathological effects of pepper (oleoresin capsicum) spray. Hong Kong Med. J. 2015, 21, 542–552. [Google Scholar] [CrossRef]
- Nirmala Grace, A.; Pandian, K. Antibacterial efficacy of aminoglycosidic antibiotics protected gold nanoparticles—A brief study. Colloids Surf. A Physicochem. Eng. Asp. 2007, 297, 63–70. [Google Scholar] [CrossRef]
- Turos, E.; Shim, J.-Y.; Wang, Y.; Greenhalgh, K.; Reddy, G.S.K.; Dickey, S.; Lim, D.V. Antibiotic-conjugated polyacrylate nanoparticles: New opportunities for development of anti-MRSA agents. Bioorganic Med. Chem. Lett. 2007, 17, 53–56. [Google Scholar] [CrossRef] [PubMed]
- Saha, B.; Bhattacharya, J.; Mukherjee, A.; Ghosh, A.; Santra, C.; Dasgupta, A.K.; Karmakar, P. In Vitro Structural and Functional Evaluation of Gold Nanoparticles Conjugated Antibiotics. Nanoscale Res. Lett. 2007, 2, 614. [Google Scholar] [CrossRef]
- Galdiero, S.; Falanga, A.; Vitiello, M.; Cantisani, M.; Marra, V.; Galdiero, M. Silver Nanoparticles as Potential Antiviral Agents. Molecules 2011, 16, 8894–8918. [Google Scholar] [CrossRef]
- Milovanovic, M.; Arsenijevic, A.; Milovanovic, J.; Kanjevac, T.; Arsenijevic, N. Chapter 14—Nanoparticles in Antiviral Therapy. In Antimicrobial Nanoarchitectonics; Grumezescu, A.M., Ed.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 383–410. [Google Scholar] [CrossRef]
- Gurunathan, S.; Qasim, M.; Choi, Y.; Do, J.T.; Park, C.; Hong, K.; Kim, J.-H.; Song, H. Antiviral Potential of Nanoparticles—Can Nanoparticles Fight against Coronaviruses? Nanomaterials 2020, 10, 1645. [Google Scholar] [CrossRef]
- Trombino, S.; Mellace, S.; Cassano, R. Solid lipid nanoparticles for antifungal drugs delivery for topical applications. Ther. Deliv. 2016, 7, 639–647. [Google Scholar] [CrossRef]
- Soliman, G.M. Nanoparticles as safe and effective delivery systems of antifungal agents: Achievements and challenges. Int. J. Pharm. 2017, 523, 15–32. [Google Scholar] [CrossRef]
- Nami, S.; Aghebati-Maleki, A.; Aghebati-Maleki, L. Current applications and prospects of nanoparticles for antifungal drug delivery. EXCLI J. 2021, 20, 562. [Google Scholar]
- Elmi, T.; Gholami, S.; Fakhar, M.; Azizi, F. A review on the use of nanoparticles in the treatment. J. Maz. Univ. Med. Sci. 2013, 23, 126–133. [Google Scholar]
- Rahul, S.; Chandrashekhar, P.; Hemant, B.; Bipinchandra, S.; Mouray, E.; Grellier, P.; Satish, P. In vitro antiparasitic activity of microbial pigments and their combination with phytosynthesized metal nanoparticles. Parasitol. Int. 2015, 64, 353–356. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Chen, D.; Pan, Y.; Qu, W.; Hao, H.; Wang, X.; Liu, Z.; Xie, S. Nanoparticles for antiparasitic drug delivery. Drug Deliv. 2019, 26, 1206–1221. [Google Scholar] [CrossRef]
- Date, A.A.; Joshi, M.D.; Patravale, V.B. Parasitic diseases: Liposomes and polymeric nanoparticles versus lipid nanoparticles. Adv. Drug Deliv. Rev. 2007, 59, 505–521. [Google Scholar] [CrossRef] [PubMed]
- Matei, A.-M.; Caruntu, C.; Tampa, M.; Georgescu, S.R.; Matei, C.; Constantin, M.M.; Constantin, T.V.; Calina, D.; Ciubotaru, D.A.; Badarau, I.A. Applications of nanosized-lipid-based drug delivery systems in wound care. Appl. Sci. 2021, 11, 4915. [Google Scholar] [CrossRef]
Genus | Species | Extract from | MIC (μg/mL) | Year of Research | Reference |
---|---|---|---|---|---|
Gram Positive | |||||
Staphylococcus | S. aureus | Capsicum frutescens | 1.2 | 2014 | [85] |
Capsicum chinense | not specified | 2018 | [86] | ||
Streptococcus | S. pyogenes | Capsicum spp. | 64–128 | 2015 | [87] |
Enterococcus | E. faecalis | Capsicum frutescens | 25 | 2014 | [85] |
Bacillus | B. subtillis | Capsicum frutescens | 25 | 2014 | [85] |
Listeria | L. monocytogenes | Capsicum spp. | not specified | 2018 | [88] |
Gram Negative | |||||
Vibrio | V. cholerae | Capsicum spp. | 100 | 2010 | [89] |
Acinetobacter | A. baumanii | Capsicum annuum L. | 64 | 2011 | [90] |
Helicobacter | H. pylori | Capsicum spp. | 25 | 2005 | [91] |
Salmonella | S. typhimurium | Capsicum chinense | not specified | 2022 | [92] |
Escherichia | E. coli | Capsicum frutescens | 5 | 2014 | [85] |
Capsicum chinese | not specified | 2018 | [86] | ||
Klebsiella | K. pneumoniae | Capsicum frutescens | 0.6 | 2014 | [85] |
Proteus | P. mirabilis | Capsicum annuum L. | 32 | 2011 | [90] |
Pseudomonas | P. aeruginosa | Capsicum frutescens | 10 | 2014 | [85] |
Capsicum spp. | not specified | 2018 | [93] |
Genus | Species | Extract from | MIC (μg/mL) | Year of Research | Reference |
---|---|---|---|---|---|
Candida | C. albicans, C. glabrata, C. tropicalis | Capsicum frutescens | 25 (MIC100) | 2014 | [85] |
Capsicum chinense | 187.5–1500 (MIC100) | 2022 | [189] | ||
Apsergillus | A. parasiticus | Capsicum chinense | 68 (MIC50) | 2020 | [190] |
381 (MIC50) | 2020 | [191] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Periferakis, A.-T.; Periferakis, A.; Periferakis, K.; Caruntu, A.; Badarau, I.A.; Savulescu-Fiedler, I.; Scheau, C.; Caruntu, C. Antimicrobial Properties of Capsaicin: Available Data and Future Research Perspectives. Nutrients 2023, 15, 4097. https://doi.org/10.3390/nu15194097
Periferakis A-T, Periferakis A, Periferakis K, Caruntu A, Badarau IA, Savulescu-Fiedler I, Scheau C, Caruntu C. Antimicrobial Properties of Capsaicin: Available Data and Future Research Perspectives. Nutrients. 2023; 15(19):4097. https://doi.org/10.3390/nu15194097
Chicago/Turabian StylePeriferakis, Aristodemos-Theodoros, Argyrios Periferakis, Konstantinos Periferakis, Ana Caruntu, Ioana Anca Badarau, Ilinca Savulescu-Fiedler, Cristian Scheau, and Constantin Caruntu. 2023. "Antimicrobial Properties of Capsaicin: Available Data and Future Research Perspectives" Nutrients 15, no. 19: 4097. https://doi.org/10.3390/nu15194097
APA StylePeriferakis, A. -T., Periferakis, A., Periferakis, K., Caruntu, A., Badarau, I. A., Savulescu-Fiedler, I., Scheau, C., & Caruntu, C. (2023). Antimicrobial Properties of Capsaicin: Available Data and Future Research Perspectives. Nutrients, 15(19), 4097. https://doi.org/10.3390/nu15194097