Sarecycline Demonstrated Reduced Activity Compared to Minocycline against Microbial Species Representing Human Gastrointestinal Microbiota
Abstract
:1. Introduction
2. Results
2.1. Effect of Sarecycline Compared to Minocycline on Gut Microbiota In Vitro
2.2. Effect of Sarecycline and Minocycline on Microbial Growth
2.2.1. Aerobic Species
2.2.2. Anaerobic Species
3. Discussion
4. Materials and Methods
4.1. Representative Gut Bacterial and Fungal Strains
4.2. Antimicrobial Susceptibility Testing
4.2.1. Anaerobic Bacteria
4.2.2. Yeasts
4.3. Aerobic Growth Curve Conditions
4.4. Anaerobic Growth Curve Conditions
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Byndloss, M.X.; Pernitzsch, S.R.; Baumler, A.J. Healthy hosts rule within: Ecological forces shaping the gut microbiota. Mucosal Immunol. 2018, 11, 1299–1305. [Google Scholar] [CrossRef] [Green Version]
- Shreiner, A.B.; Kao, J.Y.; Young, V.B. The gut microbiome in health and in disease. Curr. Opin. Gastroenterol. 2015, 31, 69–75. [Google Scholar] [CrossRef] [PubMed]
- Becker, E.; Schmidt, T.S.B.; Bengs, S.; Poveda, L.; Opitz, L.; Atrott, K.; Stanzel, C.; Biedermann, L.; Rehman, A.; Jonas, D.; et al. Effects of oral antibiotics and isotretinoin on the murine gut microbiota. Int. J. Antimicrob. Agents 2017, 50, 342–351. [Google Scholar] [CrossRef] [Green Version]
- Ferrer, M.; Mendez-Garcia, C.; Rojo, D.; Barbas, C.; Moya, A. Antibiotic use and microbiome function. Biochem. Pharmacol. 2017, 134, 114–126. [Google Scholar] [CrossRef] [PubMed]
- Zaura, E.; Brandt, B.W.; Teixeira de Mattos, M.J.; Buijs, M.J.; Caspers, M.P.; Rashid, M.U.; Weintraub, A.; Nord, C.E.; Savell, A.; Hu, Y.; et al. Same Exposure but Two Radically Different Responses to Antibiotics: Resilience of the Salivary Microbiome versus Long-Term Microbial Shifts in Feces. mBio 2015, 6, e01615–e01693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schwartz, D.J.; Langdon, A.E.; Dantas, G. Understanding the impact of antibiotic perturbation on the human microbiome. Genome Med. 2020, 12, 82. [Google Scholar] [CrossRef]
- Ejtahed, H.S.; Mohtadi-Nia, J.; Homayouni-Rad, A.; Niafar, M.; Asghari-Jafarabadi, M.; Mofid, V. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition 2012, 28, 539–543. [Google Scholar] [CrossRef]
- Tonucci, L.B.; Olbrich Dos Santos, K.M.; Licursi de Oliveira, L.; Rocha Ribeiro, S.M.; Duarte Martino, H.S. Clinical application of probiotics in type 2 diabetes mellitus: A randomized, double-blind, placebo-controlled study. Clin. Nutr. 2017, 36, 85–92. [Google Scholar] [CrossRef] [PubMed]
- Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef]
- Forslund, K.; Hildebrand, F.; Nielsen, T.; Falony, G.; Le Chatelier, E.; Sunagawa, S.; Prifti, E.; Vieira-Silva, S.; Gudmundsdottir, V.; Pedersen, H.K.; et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2015, 528, 262–266. [Google Scholar] [CrossRef] [PubMed]
- Larsen, N.; Vogensen, F.K.; van den Berg, F.W.; Nielsen, D.S.; Andreasen, A.S.; Pedersen, B.K.; Al-Soud, W.A.; Sorensen, S.J.; Hansen, L.H.; Jakobsen, M. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE 2010, 5, e9085. [Google Scholar] [CrossRef] [PubMed]
- Santacruz, A.; Collado, M.C.; Garcia-Valdes, L.; Segura, M.T.; Martin-Lagos, J.A.; Anjos, T.; Marti-Romero, M.; Lopez, R.M.; Florido, J.; Campoy, C.; et al. Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Br. J. Nutr. 2010, 104, 83–92. [Google Scholar] [CrossRef] [Green Version]
- Zupancic, M.L.; Cantarel, B.L.; Liu, Z.; Drabek, E.F.; Ryan, K.A.; Cirimotich, S.; Jones, C.; Knight, R.; Walters, W.A.; Knights, D.; et al. Analysis of the gut microbiota in the old order Amish and its relation to the metabolic syndrome. PLoS ONE 2012, 7, e43052. [Google Scholar] [CrossRef] [PubMed]
- Clarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucey, A.J.; Humphreys, M.; Hogan, A.; Hayes, P.; O’Reilly, M.; Jeffery, I.B.; Wood-Martin, R.; et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 2014, 63, 1913–1920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef] [PubMed]
- Sze, M.A.; Schloss, P.D. Looking for a Signal in the Noise: Revisiting Obesity and the Microbiome. mBio 2016, 7, e01018-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schwiertz, A.; Taras, D.; Schafer, K.; Beijer, S.; Bos, N.A.; Donus, C.; Hardt, P.D. Microbiota and SCFA in lean and overweight healthy subjects. Obesity 2010, 18, 190–195. [Google Scholar] [CrossRef]
- Kaufman, D.W.; Kelly, J.P.; Curhan, G.C.; Anderson, T.E.; Dretler, S.P.; Preminger, G.M.; Cave, D.R. Oxalobacter formigenes may reduce the risk of calcium oxalate kidney stones. J. Am. Soc. Nephrol. 2008, 19, 1197–1203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barnett, C.; Nazzal, L.; Goldfarb, D.S.; Blaser, M.J. The Presence of Oxalobacter formigenes in the Microbiome of Healthy Young Adults. J. Urol. 2016, 195, 499–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.Y.; Lee, E.; Park, Y.M.; Hong, S.J. Microbiome in the Gut-Skin Axis in Atopic Dermatitis. Allergy Asthma Immunol. Res. 2018, 10, 354–362. [Google Scholar] [CrossRef] [PubMed]
- Joossens, M.; Huys, G.; Cnockaert, M.; De Preter, V.; Verbeke, K.; Rutgeerts, P.; Vandamme, P.; Vermeire, S. Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives. Gut 2011, 60, 631–637. [Google Scholar] [CrossRef] [Green Version]
- Png, C.W.; Linden, S.K.; Gilshenan, K.S.; Zoetendal, E.G.; McSweeney, C.S.; Sly, L.I.; McGuckin, M.A.; Florin, T.H. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am. J. Gastroenterol. 2010, 105, 2420–2428. [Google Scholar] [CrossRef]
- Loubinoux, J.; Bronowicki, J.P.; Pereira, I.A.; Mougenel, J.L.; Faou, A.E. Sulfate-reducing bacteria in human feces and their association with inflammatory bowel diseases. FEMS Microbiol. Ecol. 2002, 40, 107–112. [Google Scholar] [CrossRef]
- McKee, A.M.; Kirkup, B.M.; Madgwick, M.; Fowler, W.J.; Price, C.A.; Dreger, S.A.; Ansorge, R.; Makin, K.A.; Caim, S.; Le Gall, G.; et al. Antibiotic-induced disturbances of the gut microbiota result in accelerated breast tumor growth. iScience 2021, 24, 103012. [Google Scholar] [CrossRef]
- Margolis, D.J.; Fanelli, M.; Hoffstad, O.; Lewis, J.D. Potential association between the oral tetracycline class of antimicrobials used to treat acne and inflammatory bowel disease. Am. J. Gastroenterol. 2010, 105, 2610–2616. [Google Scholar] [CrossRef]
- De Pessemier, B.; Grine, L.; Debaere, M.; Maes, A.; Paetzold, B.; Callewaert, C. Gut-Skin Axis: Current Knowledge of the Interrelationship between Microbial Dysbiosis and Skin Conditions. Microorganisms 2021, 9, 353. [Google Scholar] [CrossRef] [PubMed]
- Thompson, K.G.; Rainer, B.M.; Antonescu, C.; Florea, L.; Mongodin, E.F.; Kang, S.; Chien, A.L. Minocycline and Its Impact on Microbial Dysbiosis in the Skin and Gastrointestinal Tract of Acne Patients. Ann. Dermatol. 2020, 32, 21–30. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Kuo, S.; Shu, M.; Yu, J.; Huang, S.; Dai, A.; Two, A.; Gallo, R.L.; Huang, C.M. Staphylococcus epidermidis in the human skin microbiome mediates fermentation to inhibit the growth of Propionibacterium acnes: Implications of probiotics in acne vulgaris. Appl. Microbiol. Biotechnol. 2014, 98, 411–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jang, H.M.; Lee, K.E.; Kim, D.H. The Preventive and Curative Effects of Lactobacillus reuteri NK33 and Bifidobacterium adolescentis NK98 on Immobilization Stress-Induced Anxiety/Depression and Colitis in Mice. Nutrients 2019, 11, 819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tian, P.; O’Riordan, K.J.; Lee, Y.K.; Wang, G.; Zhao, J.; Zhang, H.; Cryan, J.F.; Chen, W. Towards a psychobiotic therapy for depression: Bifidobacterium breve CCFM1025 reverses chronic stress-induced depressive symptoms and gut microbial abnormalities in mice. Neurobiol. Stress 2020, 12, 100216. [Google Scholar] [CrossRef] [PubMed]
- Messaoudi, S.; Manai, M.; Kergourlay, G.; Prevost, H.; Connil, N.; Chobert, J.M.; Dousset, X. Lactobacillus salivarius: Bacteriocin and probiotic activity. Food Microbiol. 2013, 36, 296–304. [Google Scholar] [CrossRef]
- Batool, Z.; Lomakin, I.B.; Polikanov, Y.S.; Bunick, C.G. Sarecycline interferes with tRNA accommodation and tethers mRNA to the 70S ribosome. Proc. Natl. Acad. Sci. USA 2020, 117, 20530–20537. [Google Scholar] [CrossRef]
- Zhanel, G.; Critchley, I.; Lin, L.Y.; Alvandi, N. Microbiological Profile of Sarecycline, a Novel Targeted Spectrum Tetracycline for the Treatment of Acne Vulgaris. Antimicrob. Agents Chemother. 2019, 63, e01297-18. [Google Scholar] [CrossRef] [Green Version]
- Bunick, C.G.; Keri, J.; Tanaka, S.K.; Furey, N.; Damiani, G.; Johnson, J.L.; Grada, A. Antibacterial Mechanisms and Efficacy of Sarecycline in Animal Models of Infection and Inflammation. Antibiotics 2021, 10, 439. [Google Scholar] [CrossRef]
- Zou, L.; Mei, Z.; Guan, T.; Zhang, B.; Deng, Q. Underlying mechanisms of the effect of minocycline against Candida albicans biofilms. Exp. Ther. Med. 2021, 21, 413. [Google Scholar] [CrossRef]
- Rabah, H.; Rosa do Carmo, F.L.; Jan, G. Dairy Propionibacteria: Versatile Probiotics. Microorganisms 2017, 5, 24. [Google Scholar] [CrossRef] [Green Version]
- Yoshida, N.; Emoto, T.; Yamashita, T.; Watanabe, H.; Hayashi, T.; Tabata, T.; Hoshi, N.; Hatano, N.; Ozawa, G.; Sasaki, N.; et al. Bacteroides vulgatus and Bacteroides dorei Reduce Gut Microbial Lipopolysaccharide Production and Inhibit Atherosclerosis. Circulation 2018, 138, 2486–2498. [Google Scholar] [CrossRef]
- Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K.; et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232–236. [Google Scholar] [CrossRef]
- Mandic, A.D.; Woting, A.; Jaenicke, T.; Sander, A.; Sabrowski, W.; Rolle-Kampcyk, U.; von Bergen, M.; Blaut, M. Clostridium ramosum regulates enterochromaffin cell development and serotonin release. Sci. Rep. 2019, 9, 1177. [Google Scholar] [CrossRef] [Green Version]
- Su, L.; Nalle, S.C.; Shen, L.; Turner, E.S.; Singh, G.; Breskin, L.A.; Khramtsova, E.A.; Khramtsova, G.; Tsai, P.Y.; Fu, Y.X.; et al. TNFR2 activates MLCK-dependent tight junction dysregulation to cause apoptosis-mediated barrier loss and experimental colitis. Gastroenterology 2013, 145, 407–415. [Google Scholar] [CrossRef] [Green Version]
- Round, J.L.; Mazmanian, S.K. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl. Acad. Sci. USA 2010, 107, 12204–12209. [Google Scholar] [CrossRef] [Green Version]
- Mazmanian, S.K.; Round, J.L.; Kasper, D.L. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 2008, 453, 620–625. [Google Scholar] [CrossRef] [Green Version]
- Round, J.L.; Lee, S.M.; Li, J.; Tran, G.; Jabri, B.; Chatila, T.A.; Mazmanian, S.K. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 2011, 332, 974–977. [Google Scholar] [CrossRef] [Green Version]
- Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef] [Green Version]
- Takaishi, H.; Matsuki, T.; Nakazawa, A.; Takada, T.; Kado, S.; Asahara, T.; Kamada, N.; Sakuraba, A.; Yajima, T.; Higuchi, H.; et al. Imbalance in intestinal microflora constitution could be involved in the pathogenesis of inflammatory bowel disease. Int. J. Med. Microbiol. 2008, 298, 463–472. [Google Scholar] [CrossRef]
- Manichanh, C.; Rigottier-Gois, L.; Bonnaud, E.; Gloux, K.; Pelletier, E.; Frangeul, L.; Nalin, R.; Jarrin, C.; Chardon, P.; Marteau, P.; et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 2006, 55, 205–211. [Google Scholar] [CrossRef] [Green Version]
- Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [Green Version]
- Louis, P.; Flint, H.J. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol. Lett. 2009, 294, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef] [Green Version]
- Lukovac, S.; Belzer, C.; Pellis, L.; Keijser, B.J.; de Vos, W.M.; Montijn, R.C.; Roeselers, G. Differential modulation by Akkermansia muciniphila and Faecalibacterium prausnitzii of host peripheral lipid metabolism and histone acetylation in mouse gut organoids. mBio 2014, 5, e01438-14. [Google Scholar] [CrossRef] [Green Version]
- Park, J.H.; Kotani, T.; Konno, T.; Setiawan, J.; Kitamura, Y.; Imada, S.; Usui, Y.; Hatano, N.; Shinohara, M.; Saito, Y.; et al. Promotion of Intestinal Epithelial Cell Turnover by Commensal Bacteria: Role of Short-Chain Fatty Acids. PLoS ONE 2016, 11, e0156334. [Google Scholar] [CrossRef]
- Kelly, C.J.; Zheng, L.; Campbell, E.L.; Saeedi, B.; Scholz, C.C.; Bayless, A.J.; Wilson, K.E.; Glover, L.E.; Kominsky, D.J.; Magnuson, A.; et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 2015, 17, 662–671. [Google Scholar] [CrossRef] [Green Version]
- Miao, W.; Wu, X.; Wang, K.; Wang, W.; Wang, Y.; Li, Z.; Liu, J.; Li, L.; Peng, L. Sodium Butyrate Promotes Reassembly of Tight Junctions in Caco-2 Monolayers Involving Inhibition of MLCK/MLC2 Pathway and Phosphorylation of PKCbeta2. Int. J. Mol. Sci. 2016, 17, 1696. [Google Scholar] [CrossRef] [Green Version]
- Peng, L.; Li, Z.R.; Green, R.S.; Holzman, I.R.; Lin, J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J. Nutr. 2009, 139, 1619–1625. [Google Scholar] [CrossRef]
- Valenzano, M.C.; DiGuilio, K.; Mercado, J.; Teter, M.; To, J.; Ferraro, B.; Mixson, B.; Manley, I.; Baker, V.; Moore, B.A.; et al. Remodeling of Tight Junctions and Enhancement of Barrier Integrity of the CACO-2 Intestinal Epithelial Cell Layer by Micronutrients. PLoS ONE 2015, 10, e0133926. [Google Scholar] [CrossRef] [Green Version]
- Zheng, L.; Kelly, C.J.; Battista, K.D.; Schaefer, R.; Lanis, J.M.; Alexeev, E.E.; Wang, R.X.; Onyiah, J.C.; Kominsky, D.J.; Colgan, S.P. Microbial-Derived Butyrate Promotes Epithelial Barrier Function through IL-10 Receptor-Dependent Repression of Claudin-2. J. Immunol. 2017, 199, 2976–2984. [Google Scholar] [CrossRef] [Green Version]
- Donohoe, D.R.; Garge, N.; Zhang, X.; Sun, W.; O’Connell, T.M.; Bunger, M.K.; Bultman, S.J. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011, 13, 517–526. [Google Scholar] [CrossRef] [Green Version]
- Riviere, A.; Selak, M.; Lantin, D.; Leroy, F.; De Vuyst, L. Bifidobacteria and Butyrate-Producing Colon Bacteria: Importance and Strategies for Their Stimulation in the Human Gut. Front. Microbiol. 2016, 7, 979. [Google Scholar] [CrossRef] [Green Version]
- Cox, M.A.; Jackson, J.; Stanton, M.; Rojas-Triana, A.; Bober, L.; Laverty, M.; Yang, X.; Zhu, F.; Liu, J.; Wang, S.; et al. Short-chain fatty acids act as antiinflammatory mediators by regulating prostaglandin E(2) and cytokines. World J. Gastroenterol. 2009, 15, 5549–5557. [Google Scholar] [CrossRef]
- Mu, Q.; Kirby, J.; Reilly, C.M.; Luo, X.M. Leaky Gut As a Danger Signal for Autoimmune Diseases. Front. Immunol. 2017, 8, 598. [Google Scholar] [CrossRef] [Green Version]
- Francino, M.P. Antibiotics and the Human Gut Microbiome: Dysbioses and Accumulation of Resistances. Front. Microbiol. 2015, 6, 1543. [Google Scholar] [CrossRef] [Green Version]
- Sun, L.; Zhang, X.; Zhang, Y.; Zheng, K.; Xiang, Q.; Chen, N.; Chen, Z.; Zhang, N.; Zhu, J.; He, Q. Antibiotic-Induced Disruption of Gut Microbiota Alters Local Metabolomes and Immune Responses. Front. Cell Infect. Microbiol. 2019, 9, 99. [Google Scholar] [CrossRef]
- Knoop, K.A.; McDonald, K.G.; Kulkarni, D.H.; Newberry, R.D. Antibiotics promote inflammation through the translocation of native commensal colonic bacteria. Gut 2016, 65, 1100–1109. [Google Scholar] [CrossRef] [Green Version]
- Kiecolt-Glaser, J.K.; Wilson, S.J.; Bailey, M.L.; Andridge, R.; Peng, J.; Jaremka, L.M.; Fagundes, C.P.; Malarkey, W.B.; Laskowski, B.; Belury, M.A. Marital distress, depression, and a leaky gut: Translocation of bacterial endotoxin as a pathway to inflammation. Psychoneuroendocrinology 2018, 98, 52–60. [Google Scholar] [CrossRef]
- Stehle, J.R., Jr.; Leng, X.; Kitzman, D.W.; Nicklas, B.J.; Kritchevsky, S.B.; High, K.P. Lipopolysaccharide-binding protein, a surrogate marker of microbial translocation, is associated with physical function in healthy older adults. J. Gerontol. A Biol. Sci. Med. Sci. 2012, 67, 1212–1218. [Google Scholar] [CrossRef]
- Tulstrup, M.V.; Christensen, E.G.; Carvalho, V.; Linninge, C.; Ahrne, S.; Hojberg, O.; Licht, T.R.; Bahl, M.I. Antibiotic Treatment Affects Intestinal Permeability and Gut Microbial Composition in Wistar Rats Dependent on Antibiotic Class. PLoS ONE 2015, 10, e0144854. [Google Scholar] [CrossRef]
- Wang, F.; Schwarz, B.T.; Graham, W.V.; Wang, Y.; Su, L.; Clayburgh, D.R.; Abraham, C.; Turner, J.R. IFN-gamma-induced TNFR2 expression is required for TNF-dependent intestinal epithelial barrier dysfunction. Gastroenterology 2006, 131, 1153–1163. [Google Scholar] [CrossRef] [Green Version]
- Xavier, R.J.; Podolsky, D.K. Unravelling the pathogenesis of inflammatory bowel disease. Nature 2007, 448, 427–434. [Google Scholar] [CrossRef]
- Khaleghi, S.; Ju, J.M.; Lamba, A.; Murray, J.A. The potential utility of tight junction regulation in celiac disease: Focus on larazotide acetate. Therap. Adv. Gastroenterol. 2016, 9, 37–49. [Google Scholar] [CrossRef] [Green Version]
- Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, Y.M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [Green Version]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
- Levison, M.E. Pharmacodynamics of antimicrobial drugs. Infect. Dis. Clin. N. Am. 2004, 18, 451–465. [Google Scholar] [CrossRef]
- Levison, M.E.; Levison, J.H. Pharmacokinetics and pharmacodynamics of antibacterial agents. Infect. Dis. Clin. N. Am. 2009, 23, 791–815. [Google Scholar] [CrossRef] [Green Version]
- Bowker, K.E.; Noel, A.R.; Macgowan, A.P. Pharmacodynamics of minocycline against Staphylococcus aureus in an in vitro pharmacokinetic model. Antimicrob. Agents Chemother. 2008, 52, 4370–4373. [Google Scholar] [CrossRef] [Green Version]
- Jacob, L.; John, M.; Kalder, M.; Kostev, K. Prevalence of vulvovaginal candidiasis in gynecological practices in Germany: A retrospective study of 954,186 patients. Curr. Med. Mycol. 2018, 4, 6–11. [Google Scholar] [CrossRef]
- Almirall, L. Seysara Package Insert. Available online: https://www.almirall.us/pdf/Seysara_uspi_final_Jun2020.pdf (accessed on 12 December 2021).
- Moore, A.; Green, L.J.; Bruce, S.; Sadick, N.; Tschen, E.; Werschler, P.; Cook-Bolden, F.E.; Dhawan, S.S.; Forsha, D.; Gold, M.H.; et al. Once-Daily Oral Sarecycline 1.5 mg/kg/day Is Effective for Moderate to Severe Acne Vulgaris: Results from Two Identically Designed, Phase 3, Randomized, Double-Blind Clinical Trials. J. Drugs Dermatol. 2018, 17, 987–996. [Google Scholar] [CrossRef]
- Ben-Ami, R.; Olshtain-Pops, K.; Krieger, M.; Oren, I.; Bishara, J.; Dan, M.; Wiener-Well, Y.; Weinberger, M.; Zimhony, O.; Chowers, M.; et al. Antibiotic exposure as a risk factor for fluconazole-resistant Candida bloodstream infection. Antimicrob. Agents Chemother. 2012, 56, 2518–2523. [Google Scholar] [CrossRef] [Green Version]
- Noverr, M.C.; Huffnagle, G.B. Regulation of Candida albicans morphogenesis by fatty acid metabolites. Infect. Immun. 2004, 72, 6206–6210. [Google Scholar] [CrossRef] [Green Version]
- Kennedy, M.J.; Volz, P.A. Effect of various antibiotics on gastrointestinal colonization and dissemination by Candida albicans. Sabouraudia 1985, 23, 265–273. [Google Scholar] [CrossRef]
- Huffnagle, G.B.; Noverr, M.C. The emerging world of the fungal microbiome. Trends Microbiol. 2013, 21, 334–341. [Google Scholar] [CrossRef] [Green Version]
- King, C.H.; Desai, H.; Sylvetsky, A.C.; LoTempio, J.; Ayanyan, S.; Carrie, J.; Crandall, K.A.; Fochtman, B.C.; Gasparyan, L.; Gulzar, N.; et al. Baseline human gut microbiota profile in healthy people and standard reporting template. PLoS ONE 2019, 14, e0206484. [Google Scholar] [CrossRef] [Green Version]
- CLSI. Methods for Antimicrobial Susceptibility Testing of Anaerobic Bacteria, 7th ed.; Approved Standard; CLSI: Chicago, IL, USA, 2004; Volume 27, No. 2. [Google Scholar]
- Maier, L.; Pruteanu, M.; Kuhn, M.; Zeller, G.; Telzerow, A.; Anderson, E.E.; Brochado, A.R.; Fernandez, K.C.; Dose, H.; Mori, H.; et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature 2018, 555, 623–628. [Google Scholar] [CrossRef]
- Ejim, L.; Farha, M.A.; Falconer, S.B.; Wildenhain, J.; Coombes, B.K.; Tyers, M.; Brown, E.D.; Wright, G.D. Combinations of antibiotics and nonantibiotic drugs enhance antimicrobial efficacy. Nat. Chem. Biol. 2011, 7, 348–350. [Google Scholar] [CrossRef]
- Barbieri, J.S.; Bhate, K.; Hartnett, K.P.; Fleming-Dutra, K.E.; Margolis, D.J. Trends in Oral Antibiotic Prescription in Dermatology, 2008 to 2016. JAMA Dermatol. 2019, 155, 290–297. [Google Scholar] [CrossRef]
- Ventola, C.L. The antibiotic resistance crisis: Part 1: Causes and threats. P T 2015, 40, 277–283. [Google Scholar]
- Cui, D.; Liu, X.; Hawkey, P.; Li, H.; Wang, Q.; Mao, Z.; Sun, J. Use of and microbial resistance to antibiotics in China: A path to reducing antimicrobial resistance. J. Int. Med. Res. 2017, 45, 1768–1778. [Google Scholar] [CrossRef]
Phylum | Genus | Species | Sarecycline | Minocycline | MIC Fold Difference |
---|---|---|---|---|---|
Actinobacteria | Bifidobacterium | Bifidobacterium adolescentis | 1 | 1 | 1 |
Actinobacteria | Collinsella | Collinsella aerofaciens | 1 | 0.5 | 2 |
Actinobacteria | Eggerthella | Eggerthella lenta | 1 | 0.5 | 2 |
Actinobacteria | Actinomycetales | Propionibacterium freudenreichii | 8 | 1 | 8 |
Bacteroidetes | Bacteroides | Bacteroides caccae | 8 | 0.25 | 32 |
Bacteroidetes | Bacteroides | Bacteroides fragilis enterotoxigenic (ET) | 2 | 4 | 0.5 |
Bacteroidetes | Bacteroides | Bacteroides fragilis nontoxigenic | 1 | 0.25 | 4 |
Bacteroidetes | Bacteroides | Bacteroides ovatus | 0.5 | 0.5 | 1 |
Bacteroidetes | Bacteroides | Bacteroides thetaiotaomicron | 0.25 | 0.125 | 2 |
Bacteroidetes | Bacteroides | Bacteroides uniformis | 2 | 0.5 | 4 |
Bacteroidetes | Bacteroides | Bacteroides vulgatus | 0.125 | 0.016 | 7.8 |
Bacteroidetes | Bacteroides | Bacteroides xylanisolvens | 1 | 0.25 | 4 |
Bacteroidetes | Bacteroides | Bifidobacterium subtile Biavati | >8 | 8 | ND * |
Bacteroidetes | Odoribacter | Odoribacter splanchnicus | 8 | 4 | 2 |
Bacteroidetes | Parabacteroides | Parabacteroides distasonis | 8 | 2 | 4 |
Bacteroidetes | Parabacteroides | Parabacteroides merdae | 0.06 | 0.016 | 3.8 |
Firmicutes | Blautia | Blautia obeum | 1 | 0.5 | 2 |
Firmicutes | Clostridium | Clostridium bolteae | 4 | 0.5 | 8 |
Firmicutes | Clostridium | Erysipelatoclostridium ramosum | 2 | 0.06 | 33.3 |
Firmicutes | Clostridium | Clostridium saccharolyticum | 2 | 2 | 1 |
Firmicutes | Dorea | Dorea formicigenerans | 0.25 | 0.06 | 4.2 |
Firmicutes | Eubacterium | Eubacterium eligens | >8 | 4 | ND * |
Firmicutes | Lactobacillus | Lactobacillus paracasei | 1 | 0.25 | 4 |
Proteobacteria | Escherichia | Escherichia coli IAI1 | 16 | 8 | 2 |
Ascomycota | Candida | Candida albicans | 32 | 16 | 2 |
Ascomycota | Candida | Candida glabrata | 32 | 32 | 1 |
Ascomycota | Candida | Candida parapsilosis | 32 | 16 | 2 |
Ascomycota | Candida | Candida tropicalis | 16 | 16 | 1 |
Phylum | Genus | Species | Source |
---|---|---|---|
Bacteroidetes | Bacteroides | Bacteroides vulgatus | DSMZ 1447 |
Bacteroidetes | Bacteroides | Bacteroides uniformis | DSMZ 6597 |
Bacteroidetes | Bacteroides | Bacteroides fragilis nontoxigenic | ATCC 43858 |
Bacteroidetes | Bacteroides | Bacteroides thetaiotaomicron | DSMZ 2079 |
Bacteroidetes | Bacteroides | Bifidobacterium subtile Biavati | ATCC 27537 |
Firmicutes | Clostridium | Clostridium ramosum | DSMZ 1402 |
Actinobacteria | Bifidobacterium | Bifidobacterium adolescentis | DSMZ 20083 |
Actinobacteria | Eggerthella | Eggerthella lenta | DSMZ 2243 |
Firmicutes | Clostridium | Clostridium bolteae | DSMZ 15670 |
Bacteroidetes | Bacteroides | Bacteroides fragilis enterotoxigenic (ET) | ATCC 43860 |
Firmicutes | Clostridium | Clostridium saccharolyticum | DSMZ 2544 |
Firmicutes | Lactobacillus | Lactobacillus paracasei | DSMZ 5622 |
Bacteroidetes | Bacteroides | Bacteroides caccae | DSMZ 19024 |
Bacteroidetes | Bacteroides | Bacteroides ovatus | DSMZ 1896 |
Bacteroidetes | Bacteroides | Bacteroides xylanisolvens | DSMZ 18836 |
Firmicutes | Blautia | Blautia obeum | DSMZ 25238 |
Bacteroidetes | Parabacteroides | Parabacteroides merdae | DSMZ 19495 |
Actinobacteria | Collinsella | Collinsella aerofaciens | DSMZ 3979 |
Actinobacteria | Actinomycetales | Propionibacterium freudenreichii | CMM |
Bacteroidetes | Parabacteroides | Parabacteroides distasonis | DSMZ 20701 |
Firmicutes | Eubacterium | Eubacterium eligens | DSMZ 3376 |
Firmicutes | Dorea | Dorea formicigenerans | DSMZ 3992 |
Proteobacteria | Escherichia | Escherichia coli IAI1 | CMM |
Bacteroidetes | Odoribacter | Odoribacter splanchnicus | DSMZ 20712 |
Ascomycota | Candida | Candida albicans | CMM |
Ascomycota | Candida | Candida tropicalis | CMM |
Ascomycota | Candida | Candida parapsilosis | CMM |
Ascomycota | Candida | Candida glabrata | CMM |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Ghannoum, M.A.; Long, L.; Bunick, C.G.; Del Rosso, J.Q.; Gamal, A.; Tyring, S.K.; McCormick, T.S.; Grada, A. Sarecycline Demonstrated Reduced Activity Compared to Minocycline against Microbial Species Representing Human Gastrointestinal Microbiota. Antibiotics 2022, 11, 324. https://doi.org/10.3390/antibiotics11030324
Ghannoum MA, Long L, Bunick CG, Del Rosso JQ, Gamal A, Tyring SK, McCormick TS, Grada A. Sarecycline Demonstrated Reduced Activity Compared to Minocycline against Microbial Species Representing Human Gastrointestinal Microbiota. Antibiotics. 2022; 11(3):324. https://doi.org/10.3390/antibiotics11030324
Chicago/Turabian StyleGhannoum, Mahmoud A., Lisa Long, Christopher G. Bunick, James Q. Del Rosso, Ahmed Gamal, Stephen K. Tyring, Thomas S. McCormick, and Ayman Grada. 2022. "Sarecycline Demonstrated Reduced Activity Compared to Minocycline against Microbial Species Representing Human Gastrointestinal Microbiota" Antibiotics 11, no. 3: 324. https://doi.org/10.3390/antibiotics11030324
APA StyleGhannoum, M. A., Long, L., Bunick, C. G., Del Rosso, J. Q., Gamal, A., Tyring, S. K., McCormick, T. S., & Grada, A. (2022). Sarecycline Demonstrated Reduced Activity Compared to Minocycline against Microbial Species Representing Human Gastrointestinal Microbiota. Antibiotics, 11(3), 324. https://doi.org/10.3390/antibiotics11030324