1
|
Wang L, Geng Z, Liu Y, Cao L, Liu Y, Zhang H, Bi Y, Lu J. Multi-Modal Design, Synthesis, and Biological Evaluation of Novel Fusidic Acid Derivatives. Molecules 2025; 30:1983. [PMID: 40363790 PMCID: PMC12073777 DOI: 10.3390/molecules30091983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Fusidic acid (FA), a tetracyclic triterpenoid, has been approved to treat methicillin-resistant Staphylococcus aureus (MRSA) infections. However, there are few reports about FA derivatives with high efficacy superior to FA, manifesting the difficulty of discovering the derivatives based on experience-based drug design. In this study, we employed a stepwise method to discover novel FA derivatives. First, molecular dynamics (MD) simulations were performed to identify the molecular mechanism of FA against elongation factor G (EF-G) and drug resistance. Then, we utilized a scaffold decorator to design novel FA derivatives at the 3- and 21-positions of FA. The ligand-based and structure-based screening models, including Chemprop and RTMScore, were employed to identify promising hits from the generated set. Ten generated FA derivatives with high efficacy in the Chemprop and RTMScore models were synthesized for in vitro testing. Compounds 4 and 10 demonstrated a 2-fold increase in potency against MRSA strains compared to FA. This study highlights the significant impact of AI-based methods on the design of novel FA derivatives with drug efficacy, which provides a new approach for drug discovery.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yi Bi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (L.W.); (Z.G.); (Y.L.); (L.C.); (Y.L.); (H.Z.)
| | - Jing Lu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (L.W.); (Z.G.); (Y.L.); (L.C.); (Y.L.); (H.Z.)
| |
Collapse
|
2
|
Mukherjee S, Chakravarty S, Haldar J. Revitalizing Antibiotics with Macromolecular Engineering: Tackling Gram-Negative Superbugs and Mixed Species Bacterial Biofilm Infections In Vivo. Biomacromolecules 2025; 26:2211-2226. [PMID: 40040432 DOI: 10.1021/acs.biomac.4c01520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
The escalating prevalence of multidrug-resistant Gram-negative pathogens, coupled with dwindling antibiotic development, has created a critical void in the clinical pipeline. This alarming issue is exacerbated by the formation of biofilms by these superbugs and their frequent coexistence in mixed-species biofilms, conferring extreme antibiotic tolerance. Herein, we present an amphiphilic cationic macromolecule, ACM-AHex, as an innovative antibiotic adjuvant to rejuvenate and repurpose resistant antibiotics, for instance, rifampicin, fusidic acid, erythromycin, and chloramphenicol. ACM-AHex mildly perturbs the bacterial membrane, enhancing antibiotic permeability, hampers efflux machinery, and produces reactive oxygen species, resulting in a remarkable 64-1024-fold potentiation in antibacterial activity. The macromolecule reduces bacterial virulence and macromolecule-drug cocktail significantly eradicate both mono- and multispecies bacterial biofilms, achieving >99.9% bacterial reduction in the murine biofilm infection model. Demonstrating potent biocompatibility across multiple administration routes, ACM-AHex offers a promising strategy to restore obsolete antibiotics and combat recalcitrant Gram-negative biofilm-associated infections, advocating for further clinical evaluation as a next-generation macromolecular antibiotic adjuvant.
Collapse
Affiliation(s)
- Sudip Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Sayan Chakravarty
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| |
Collapse
|
3
|
Kim SJ, Ali MS, Kang HS, Moon BY, Hwang YJ, Yoon SS, Park SC, Lim SK. Characterization of fusidic acid-resistant Staphylococcus aureus isolated from food animals during 2010-2021 in South Korea. Int J Food Microbiol 2025; 430:111026. [PMID: 39731987 DOI: 10.1016/j.ijfoodmicro.2024.111026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/15/2024] [Accepted: 12/15/2024] [Indexed: 12/30/2024]
Abstract
Livestock-associated fusidic acid-resistant Staphylococcus aureus (FRSA) is frequently linked to global public health hazards. This study aimed to ascertain the prevalence and molecular characteristics of FRSA isolated from food animal products in South Korea from 2010 to 2021. We obtained a total of 3980 S. aureus isolates from cattle carcasses (n = 482), pig carcasses (n = 1531), and chicken carcasses (n = 1967). The isolates were evaluated for antimicrobial susceptibility using the broth microdilution method. Antimicrobial resistance genes, spa types, sequence types (STs), and Staphylococcus cassette chromosome mec (SCCmec) types were determined by polymerase chain reaction (PCR) and sequencing analysis. In total, 187 isolates (4.7 %) demonstrated resistance to fusidic acid, with the maximum recovered from cattle (16.2 %), followed by pigs (6.5 %) and chickens (0.5 %). In addition, the majority of the isolates showed resistance to penicillin (86.6 %), while comparatively low resistance rates (7-13.9 %) were observed for erythromycin, gentamicin, kanamycin, and tetracycline. Moreover, multidrug resistance (MDR) comprised 8.6 % (16/187) of the isolates. Among the fusidic acid resistance determinants, the fusA mutation was the highest, containing 54 % (101/187), followed by fusC (29.4 %, 55/187) and fusB (15.5 %, 29/187). A high level of resistance regarding the substitution of L461K in the fusA gene was identified in 97 % of isolates. In addition, the most commonly detected resistance patterns include penicillin (87.1 %, 88/101) among the FRSA. The nucleotide sequencing analysis showed that all 29 fusB-carrying isolates possess the structural gene blaZ of the bla operon and the insertion sequences orf152, orf170, IS257, and orf152. In total, 21 spa types were found, where t126 was detected the most (81.2 %, 82/101) in fusA, followed by t127 (81.8 %, 45/55) in fusC, and t189 (27.6 %, 8/29) in fusB. Furthermore, all t002 harboring fusC were detected as ST5-MRSA-SCCmecII clones. This is the first report of fusA and fusB carrying S. aureus and linkage fusB and blaZ genes in FRSA isolated from food animal products. Taken together, the FRSA in food animals with different resistance determinants and spa types could pose a threat to public health.
Collapse
Affiliation(s)
- Su-Jeong Kim
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Md Sekendar Ali
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Hee-Seung Kang
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Bo-Youn Moon
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Yu-Jeong Hwang
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Soon-Seek Yoon
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, Institute for Veterinary Biomedical Science, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Suk-Kyung Lim
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea.
| |
Collapse
|
4
|
Leitão MM, Gonçalves ASC, Moreira J, Fernandes C, Borges F, Simões M, Borges A. Unravelling the potential of natural chelating agents in the control of Staphylococcus aureus and Pseudomonas aeruginosa biofilms. Eur J Med Chem 2025; 283:117163. [PMID: 39700872 DOI: 10.1016/j.ejmech.2024.117163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/11/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Iron is essential for the formation, maturation and dispersal of bacterial biofilms, playing a crucial role in the physiological and metabolic functions of bacteria as well as in the regulation of virulence. Limited availability of iron can impair the formation of robust biofilms by altering cellular motility, hydrophobicity and protein composition of the bacterial surface. In this study, the antibiofilm activity of two natural iron chelating agents, kojic acid (5-hydroxy-2-hydroxymethyl-4H-pyran-4-one) and maltol (3-hydroxy-2-methyl-4-pyrone), were investigated against Staphylococcus aureus and Pseudomonas aeruginosa. In addition, the ability of these 2-hydroxy-4-pyrone derivatives in preventing and eradicating S. aureus and P. aeruginosa biofilms through the enhancement of the efficacy of two antibiotics (tobramycin and ciprofloxacin) was explored. The iron binding capacity of the kojic acid and maltol was confirmed by their affinity for iron (III) which was found to be about 90 %, comparable to the regular chelating agent ethylenediaminetetraacetic acid (EDTA, 89 %). The antibiofilm efficacy of 2-hydroxy-4-pyrone derivatives, alone and in combination with antibiotics, was evaluated by measuring the total biomass, metabolic activity, and culturability of biofilm cells. Furthermore, their impact on the membrane integrity of S. aureus biofilm cells was investigated using flow cytometry and epifluorescence microscopy with propidium iodide staining. It was also examined the ability of 2-hydroxy-4-pyrone derivatives and 2-hydroxy-4-pyrone derivate-antibiotic dual-combinations in inhibiting the production of virulence factors (total proteases, lipases, gelatinases and siderophores) by S. aureus. Regarding biofilm formation, the results showed that 2-hydroxy-4-pyrone derivatives alone reduced the metabolic activity of S. aureus biofilm cells by over 40 %. When combined with tobramycin, a 2-log (CFU cm-2) reduction in S. aureus biofilm cells was observed. Moreover, the combination of maltol and kojic acid with ciprofloxacin prevented P. aeruginosa biomass production by 60 %, compared to 36 % with ciprofloxacin alone. In pre-established S. aureus and P. aeruginosa biofilms, selected compounds reduced the metabolic activity by over 75 %, and a 3-log (CFU cm-2) reduction in the culturability of biofilm cells was noted when kojic acid and maltol were combined with antibiotics. Moreover, 2-hydroxy-4-pyrone derivatives alone and in combination with tobramycin, damaged the cell membranes of pre-established biofilms and completely inhibited total proteases production. Despite the increasing of reactive oxygen species production caused by the cellular treatment of maltol, both 2-hydroxy-4-pyrone derivatives showed good safe profile when tested in human hepatocarcinoma (HepG2) cells. The pre-treatment of HepG2 cells with both compounds was crucial to prevent the cellular damage caused by iron (III). This study demonstrates for the first time that the selected 2-hydroxy-4-pyrone derivatives significantly enhance the antibiofilm activity of tested antibiotics against S. aureus and P. aeruginosa, highlighting their potential as antibiotic adjuvants in preventing and eradicating biofilm-related infections.
Collapse
Affiliation(s)
- Miguel M Leitão
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007 Porto, Portugal
| | - Ariana S C Gonçalves
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; Environmental Health Department, Portuguese National Health Institute Doutor Ricardo Jorge, Porto, Portugal
| | - Joana Moreira
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007 Porto, Portugal
| | - Carlos Fernandes
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007 Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007 Porto, Portugal
| | - Manuel Simões
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; DEQ-Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal
| | - Anabela Borges
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; DEQ-Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal.
| |
Collapse
|
5
|
Yook G, Nam J, Jo Y, Yoon H, Yang D. Metabolic engineering approaches for the biosynthesis of antibiotics. Microb Cell Fact 2025; 24:35. [PMID: 39891166 PMCID: PMC11786382 DOI: 10.1186/s12934-024-02628-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/18/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Antibiotics have been saving countless lives from deadly infectious diseases, which we now often take for granted. However, we are currently witnessing a significant rise in the emergence of multidrug-resistant (MDR) bacteria, making these infections increasingly difficult to treat in hospitals. MAIN TEXT The discovery and development of new antibiotic has slowed, largely due to reduced profitability, as antibiotics often lose effectiveness quickly as pathogenic bacteria evolve into MDR strains. To address this challenge, metabolic engineering has recently become crucial in developing efficient enzymes and cell factories capable of producing both existing antibiotics and a wide range of new derivatives and analogs. In this paper, we review recent tools and strategies in metabolic engineering and synthetic biology for antibiotic discovery and the efficient production of antibiotics, their derivatives, and analogs, along with representative examples. CONCLUSION These metabolic engineering and synthetic biology strategies offer promising potential to revitalize the discovery and development of new antibiotics, providing renewed hope in humanity's fight against MDR pathogenic bacteria.
Collapse
Affiliation(s)
- Geunsoo Yook
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jiwoo Nam
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yeonseo Jo
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunji Yoon
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Dongsoo Yang
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
6
|
Kang YS, Silva SC, Smith K, Sumida K, Wang Y, Chiaraviglio L, Donthiri RR, Aljahdali AZ, Kirby JE, O’Doherty GA. Exploration of the Fusidic Acid Structure Activity Space for Antibiotic Activity. Molecules 2025; 30:465. [PMID: 39942570 PMCID: PMC11820832 DOI: 10.3390/molecules30030465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/16/2025] Open
Abstract
Fusidic acid is a translation inhibitor with activity against major Gram-positive bacterial pathogens such as S. aureus. However, its activity against Gram-negatives is poor based on an inability to access its cytoplasmic target in these organisms. Opportunities for functionalization of the fusidic acid scaffold to enhance activity against Gram-negative pathogens have not been explored. Using an activity-guided synthetic strategy, the tolerance of the tetracyclic natural product to derivatization at the A- and C-rings and its carboxylic acid side chain was explored with the goal of enhancing its activity spectrum and pharmacological properties. All side-chain carboxylic acid esters were inactive. Oxidation of the C-ring alcohol and oxime were not tolerated either. A number of esters of the A-ring alcohol retained modest activity against Gram-positive bacteria and were informative for future activity-guided studies. For the A-ring esters, differences in antibacterial activity relative to inhibitory activity in a ribosome in vitro translation assay suggested the possibility of a pro-druglike effect for the fusidic acid pyrazine-2-carboxylate. This study furthers the understanding of the activity of the fusidic acid scaffold against Gram-positive bacteria. These results suggest promise for future modification of the A-ring alcohol of fusidic acid in the advancement of its antibiotic properties.
Collapse
Affiliation(s)
- Yoon-Suk Kang
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA; (Y.-S.K.); (K.S.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Simone C. Silva
- Department of Chemistry, Northeastern University, Boston, MA 02115, USA; (S.C.S.); (K.S.); (Y.W.)
| | - Kenneth Smith
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA; (Y.-S.K.); (K.S.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Krissty Sumida
- Department of Chemistry, Northeastern University, Boston, MA 02115, USA; (S.C.S.); (K.S.); (Y.W.)
| | - Yuhan Wang
- Department of Chemistry, Northeastern University, Boston, MA 02115, USA; (S.C.S.); (K.S.); (Y.W.)
| | - Lucius Chiaraviglio
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA; (Y.-S.K.); (K.S.)
| | | | - Alhanouf Z. Aljahdali
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA; (Y.-S.K.); (K.S.)
- Harvard Medical School, Boston, MA 02115, USA
- Department of Chemistry, Northeastern University, Boston, MA 02115, USA; (S.C.S.); (K.S.); (Y.W.)
| | - James E. Kirby
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA; (Y.-S.K.); (K.S.)
- Harvard Medical School, Boston, MA 02115, USA
| | - George A. O’Doherty
- Department of Chemistry, Northeastern University, Boston, MA 02115, USA; (S.C.S.); (K.S.); (Y.W.)
| |
Collapse
|
7
|
Luo D, Ma J, Xie W, Wang Z. Metabolic analysis of the mode of action and mode of resistance of fusidic acid against Staphylococcus aureus. FEMS Microbiol Lett 2025; 372:fnaf011. [PMID: 39890598 DOI: 10.1093/femsle/fnaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 01/11/2025] [Accepted: 01/18/2025] [Indexed: 02/03/2025] Open
Abstract
Understanding bacterial responses to antibiotics is essential for identifying resistance mechanisms and developing novel therapies. This study evaluated the resistance of Staphylococcus aureus (S. aureus) to fusidic acid (FD) in 100 patients with skin and soft tissue infections (SSTIs), revealing susceptibility to FD despite resistance to other antibiotics. Through adaptive laboratory evolution, we developed a highly FD-resistant strain, E10, and identified three gene mutations (fusA, BPENGOFF-00211, and rplF) using whole-genome sequencing. The fusA mutation was the primary contributor to resistance. Furthermore, the evolved fusA mutant strain (H457Y) displayed impaired coagulation function and reduced growth rates. We also analyzed the metabolomic profiles of ancestral ATCC 25923 and evolved E10 strains, both treated and untreated with FD, revealing that the fusA gene can independently induce metabolic reprogramming. These changes primarily impacted pathways involved in central carbon metabolism, nucleotide metabolism, and amino acid synthesis. This study highlights the complexity of FD resistance in S. aureus and offers insights into the metabolic pathways associated with antibiotic resistance.
Collapse
Affiliation(s)
- Dan Luo
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of animal science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
- Collaborative Innovation Center of Agri-Seeds, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Juanjuan Ma
- Department of Clinical Laboratory, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, China
| | - Weile Xie
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of animal science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
- Collaborative Innovation Center of Agri-Seeds, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhe Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of animal science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
- Collaborative Innovation Center of Agri-Seeds, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
8
|
Chingizova EA, Yurchenko EA, Starnovskaya SS, Chingizov AR, Kuzmich AS, Pislyagin EA, Vasilchenko AS, Poshvina DV, Shilovsky GA, Dibrova DV, Aminin DL, Yurchenko AN. Flavuside B exhibits antioxidant and anti-inflammatory properties in Staphylococcus aureus infected skin wound and affect the expression of genes controlling bacterial quorum sensing. J Appl Microbiol 2025; 136:lxae318. [PMID: 39749841 DOI: 10.1093/jambio/lxae318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/15/2024] [Accepted: 12/31/2024] [Indexed: 01/04/2025]
Abstract
AIMS The aim of this study was to evaluate the antioxidant and anti-inflammatory effects of marine fungal cerebroside flavuside B (FlaB) on Staphylococcus aureus-infected keratinocytes in in vitro skin wounds and to identify FlaB targets in bacterial and human cells. METHODS AND RESULTS A combination of enzyme-linked immunosorbent assay (ELISA), plate spectrofluorimetry, and flow cytometry with fluorescence dye staining, scratch assay, and real-time cell imaging techniques was used to investigate the effects of FlaB on S. aureus-infected HaCaT keratinocytes. FlaB decreased reactive oxygen species levels, nitrite oxide levels, and TNF-α and IL-18 release in S. aureus-infected HaCaT cells. FlaB reversed the inhibition of HaCaT cell proliferation caused by S. aureus infection. FlaB significantly increased keratinocyte migration and wound healing in an in vitro S. aureus-infected wound skin model. Using real-time qPCR, we found that FlaB caused a 1.7-fold reduction in agrA expression, which controls quorum sensing system in S. aureus. Bioinformatics analysis and molecular docking, together with experimental data, suggest that FlaB targets the pro/antioxidant defense system in human cells. CONCLUSIONS Thus, FlaB can play a dual role as an antibacterial and pro/antioxidant machinery modulator, providing an observable positive effect in S. aureus-infected in vitro skin wounds. Staphylococcal sortase A enzyme and Arg systems are the targets of FlaB in bacterial cells. Nrf2/Bach1 dependent pro/antioxidant defense system is a target of FlaB in human cells. Some suggestions have also been made regarding the biological role of this marine fungal metabolite and its therapeutic possibilities.
Collapse
Affiliation(s)
- Ekaterina A Chingizova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia
| | - Ekaterina A Yurchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia
| | - Sofya S Starnovskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia
| | - Artur R Chingizov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia
| | - Aleksandra S Kuzmich
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia
| | - Evgeny A Pislyagin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia
| | - Alexey S Vasilchenko
- Laboratory of Antimicrobial Resistance, Institute of Environmental and Agricultural Biology (X-BIO), T6 Volodarskogo St., yumen State University, Tyumen 625003, Russia
| | - Darya V Poshvina
- Laboratory of Antimicrobial Resistance, Institute of Environmental and Agricultural Biology (X-BIO), T6 Volodarskogo St., yumen State University, Tyumen 625003, Russia
| | - Gregory A Shilovsky
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1 build.40 Leninskie Gory, Moscow 119234, Russia
- Faculty of Biology, Lomonosov Moscow State University, 1 build.12 Leninskie Gory, Moscow 119234, Russia
- Institute for Information Transmission Problems, Russian Academy of Sciences (Kharkevich Institute), 19 build.1 Bolshoy Karetny per., Moscow 127051, Russia
| | - Daria V Dibrova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1 build.40 Leninskie Gory, Moscow 119234, Russia
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 1 build.73 Leninskie Gory, Moscow 119991, Russia
| | - Dmitry L Aminin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia
| | - Anton N Yurchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia
| |
Collapse
|
9
|
Molasky NMR, Zhang Z, Gillespie JR, Domagala J, Reyna D, Lipka E, Fan E, Buckner FS. A novel methionyl-tRNA synthetase inhibitor targeting gram-positive bacterial pathogens. Antimicrob Agents Chemother 2024; 68:e0074524. [PMID: 39470194 PMCID: PMC11619354 DOI: 10.1128/aac.00745-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 09/15/2024] [Indexed: 10/30/2024] Open
Abstract
New antibiotics are needed to treat gram-positive bacterial pathogens. MRS-2541 is a novel inhibitor of methionyl-tRNA synthetase with selective activity against gram-positive bacteria. The minimum inhibitory concentrations (MICs) against Staphylococcus aureus, Streptococcus pyogenes, and Enterococcus species range from 0.063 to 0.5 µg/mL. Given orally to mice at 50 mg/kg every 8 hours, MRS-2541 shows sustained plasma levels well above these MICs. In the mouse thigh infection model, MRS-2541 decreased methicillin-resistant Staphylococcus aureus and Streptococcus pyogenes bacterial loads to the same degree as linezolid. MRS-2541 is a promising new antibiotic for development against skin and soft tissue infections.
Collapse
Affiliation(s)
- Nora M. R. Molasky
- Center for Emerging and Re-emerging Infectious Diseases, Department of Medicine, Division of Allergy and Infectious Disease, University of Washington, Seattle, Washington, USA
| | - Zhongsheng Zhang
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - J. Robert Gillespie
- Center for Emerging and Re-emerging Infectious Diseases, Department of Medicine, Division of Allergy and Infectious Disease, University of Washington, Seattle, Washington, USA
| | - John Domagala
- Therapeutic Systems Research Laboratories, Inc., Ann Arbor, Michigan, USA
| | - Dawn Reyna
- Therapeutic Systems Research Laboratories, Inc., Ann Arbor, Michigan, USA
| | - Elke Lipka
- Therapeutic Systems Research Laboratories, Inc., Ann Arbor, Michigan, USA
| | - Erkang Fan
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Frederick S. Buckner
- Center for Emerging and Re-emerging Infectious Diseases, Department of Medicine, Division of Allergy and Infectious Disease, University of Washington, Seattle, Washington, USA
| |
Collapse
|
10
|
Huzum B, Aprotosoaie AC, Alexa O, Sîrbu PD, Puha B, Veliceasa B, Huzum RM. Antimicrobials in Orthopedic Infections: Overview of Clinical Perspective and Microbial Resistance. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1988. [PMID: 39768868 PMCID: PMC11728363 DOI: 10.3390/medicina60121988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Orthopedic infections are challenging pathologies that impose a heavy burden on patients and the healthcare system. Antimicrobial therapy is a critical component of the successful management of orthopedic infections, but its effectiveness depends on patient-, surgery-, drug-, and hospital-related factors. The dramatic increase in the emergence of multidrug-resistant microbial strains necessitates new clinical approaches in order to prevent or limit this phenomenon and to ensure a favorable therapeutic outcome. The present paper reviews the currently available antimicrobial strategies in the management of orthopedic infections, highlighting their clinical use related to the occurrence of microbial resistance. Some approaches for reducing antibiotic resistance emergence in orthopedics are also presented. The use of antibiotics tailored to the microorganism's sensitivity profile, patient factors, and pharmacokinetic profile in terms of monotherapy or combinations, the understanding of microbial pathogenicity and resistance patterns, strict control measures in healthcare facilities, the development of new antimicrobial therapies (drugs, devices, technologies), and patient education for improving compliance and tolerance are some of the most important tools for overcoming microbial resistance.
Collapse
Affiliation(s)
- Bogdan Huzum
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Ana Clara Aprotosoaie
- Faculty of Pharmacy “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania
| | - Ovidiu Alexa
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Paul Dan Sîrbu
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Bogdan Puha
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Bogdan Veliceasa
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Riana Maria Huzum
- Department of Radiology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania;
| |
Collapse
|
11
|
Abdelmassih MM, Ismail MM, Kashef MT, Essam T. Repurposing fusidic acid as an antimicrobial against enterococci with a low probability of resistance development. Int Microbiol 2024; 27:1807-1819. [PMID: 38532184 PMCID: PMC11611940 DOI: 10.1007/s10123-024-00506-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/26/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024]
Abstract
Drug repurposing constitutes a strategy to combat antimicrobial resistance, by using agents with known safety, pharmacokinetics, and pharmacodynamics. Previous studies have implemented new fusidic acid (FA) front-loading-dose regimens, allowing higher serum levels than those achievable with ordinary doses. As susceptibility breakpoints are affected by serum level, we evaluated the repurposing of FA as an antimicrobial product against enterococci. FA minimum inhibitory concentrations (MICs) against standard enterococci strains; Enterococcus faecalis ATCC 29212 and Enterococcus faecium ATCC 27270 were 2 and 4 µg/mL, respectively. The MIC against 98 enterococcal clinical isolates was ≤ 8 µg/mL; all would be susceptible if categorized according to recalculated breakpoints (≥ 16 µg/mL), based on the serum level achieved using the front-loading regimen. FA administration in vivo, using the BALB/c mouse infection model, significantly reduced bacterial burden by two to three log10 units in the liver and spleen of mice infected with vancomycin-susceptible and -resistant strains. Exposure of the standard enterococcal strains to increasing, but not fixed, FA concentrations resulted in resistant strains (MIC = 128 µg/mL), with thicker cell walls and slower growth rates. Only one mutation (M651I) was detected in the fusA gene of the resistant strain derived from serial passage of E. faecium ATCC 27270, which was retained in the revertant strain after passage in the FA-free medium. In conclusion, FA can be repurposed as an antimicrobial drug against enterococci with a low probability of mutational resistance development, and can be employed for treatment of infections attributable to vancomycin-resistant enterococci.
Collapse
Affiliation(s)
- Mark M Abdelmassih
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Maha M Ismail
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Mona T Kashef
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Tamer Essam
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
12
|
Brdová D, Ruml T, Viktorová J. Mechanism of staphylococcal resistance to clinically relevant antibiotics. Drug Resist Updat 2024; 77:101147. [PMID: 39236354 DOI: 10.1016/j.drup.2024.101147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Staphylococcus aureus, a notorious pathogen with versatile virulence, poses a significant challenge to current antibiotic treatments due to its ability to develop resistance mechanisms against a variety of clinically relevant antibiotics. In this comprehensive review, we carefully dissect the resistance mechanisms employed by S. aureus against various antibiotics commonly used in clinical settings. The article navigates through intricate molecular pathways, elucidating the mechanisms by which S. aureus evades the therapeutic efficacy of antibiotics, such as β-lactams, vancomycin, daptomycin, linezolid, etc. Each antibiotic is scrutinised for its mechanism of action, impact on bacterial physiology, and the corresponding resistance strategies adopted by S. aureus. By synthesising the knowledge surrounding these resistance mechanisms, this review aims to serve as a comprehensive resource that provides a foundation for the development of innovative therapeutic strategies and alternative treatments for S. aureus infections. Understanding the evolving landscape of antibiotic resistance is imperative for devising effective countermeasures in the battle against this formidable pathogen.
Collapse
Affiliation(s)
- Daniela Brdová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| | - Jitka Viktorová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| |
Collapse
|
13
|
Toit SAD, Rip D. Exploring the genetic variability, virulence factors, and antibiotic resistance of Listeria monocytogenes from fresh produce, ready-to-eat hummus, and food-processing environments. J Food Sci 2024; 89:6916-6945. [PMID: 39327637 DOI: 10.1111/1750-3841.17399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/08/2024] [Accepted: 09/01/2024] [Indexed: 09/28/2024]
Abstract
Listeria monocytogenes is ubiquitous in nature and persistent in food-processing facilities, farms, retail stores, and home and restaurant kitchens. Current research suggests ready-to-eat (RTE) products (including RTE hummus and fresh produce) to be of increasing interest and concern. These foods are typically stored at refrigeration temperatures suited to the survival of L. monocytogenes and are consumed without further processing. Since L. monocytogenes is ubiquitous in agricultural environments, the cultivation of fresh produce predisposes it to contamination. The contamination of RTE foods originates either from raw ingredients or, more commonly, from cross-contamination within food-processing facilities. Research on the food-processing environment has been recommended to reduce the incidence of L. monocytogenes in foods. The consumption of contaminated foods by immunocompromised individuals causes invasive listeriosis, with a 20% to 30% fatality rate despite treatment. The emergence of antibiotic-resistant strains has reduced the effectiveness of modern medicine and may increase morbidity and mortality. Without epidemiological surveillance and identifying trends in disease determinants, no action can be taken to improve food safety and mitigate the risk of such outbreaks.
Collapse
Affiliation(s)
- Samantha Anne du Toit
- Department of Food Science, Stellenbosch University Matieland, Stellenbosch, South Africa
| | - Diane Rip
- Department of Food Science, Stellenbosch University Matieland, Stellenbosch, South Africa
| |
Collapse
|
14
|
Wood GE, Lee JW, Peramuna T, Wendt KL, Kim CM, Aguila LKT, Calderon CL, Cichewicz RH. The fungal natural product fusidic acid demonstrates potent activity against Mycoplasma genitalium. Antimicrob Agents Chemother 2024; 68:e0100624. [PMID: 39207152 PMCID: PMC11459954 DOI: 10.1128/aac.01006-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Antimicrobial resistance is extremely common in Mycoplasma genitalium, a frequent cause of urethritis in men and cervicitis, vaginitis, and pelvic inflammatory disease in women. Treatment of M. genitalium infections is difficult due to intrinsic and acquired resistance to many antibiotic classes. We undertook a program to identify novel antimicrobials with activity against M. genitalium from fungal natural products. Extracts of Ramularia coccinea contained a molecule with potent activity that was subsequently identified as fusidic acid, a fusidane-type antibiotic that has been in clinical use for decades outside the United States. We found that minimum inhibitory concentrations of fusidic acid ranged from 0.31 to 4 µg/mL among 17 M. genitalium strains including laboratory-passaged and low-passage clinical isolates. Time-kill data indicate that bactericidal killing occurs when M. genitalium is exposed to ≥10 µg/mL for 48 h, comparing favorably to serum concentrations obtained from typical loading dose regimens. Resistance to fusidic acid was associated with mutations in fusA consistent with the known mechanism of action in which fusidic acid inhibits protein synthesis by binding to elongation factor G. Interestingly, no mutants resistant to >10 µg/mL fusidic acid were obtained and a resistant strain containing a F435Y mutation in FusA was impaired for growth in vitro. These data suggest that fusidic acid may be a promising option for the treatment of M. genitalium infections.
Collapse
Affiliation(s)
- Gwendolyn E. Wood
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Jin Woo Lee
- College of Pharmacy, Duksung Women’s University, Seoul, Republic of Korea
| | - Thilini Peramuna
- Natural Products Discovery Group, Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, Institute for Natural Products Applications and Research Technologies, University of Oklahoma, Norman, Oklahoma, USA
| | - Karen L. Wendt
- Natural Products Discovery Group, Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, Institute for Natural Products Applications and Research Technologies, University of Oklahoma, Norman, Oklahoma, USA
| | - Caroline M. Kim
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Laarni Kendra T. Aguila
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Claire L. Calderon
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Robert H. Cichewicz
- Natural Products Discovery Group, Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, Institute for Natural Products Applications and Research Technologies, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
15
|
Shulenina OV, Sukhanova EA, Yarovoy BF, Tolstyko EA, Konevega AL, Paleskava A. The Antibacterial Activity of Yeasts from Unique Biocenoses. Acta Naturae 2024; 16:95-104. [PMID: 39877005 PMCID: PMC11771846 DOI: 10.32607/actanaturae.27527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/16/2024] [Indexed: 01/31/2025] Open
Abstract
The replenishment of our stock of substances that possess a therapeutic potential is an important objective in modern biomedicine. Despite the important advances achieved in chemical synthesis, the natural diversity of organisms and microorganisms remains an important source of biologically active compounds. Here, we report the results of our study of a unique collection containing more than 3,000 samples of yeasts found on the Kamchatka Peninsula, the Kuril Islands, and Sakhalin Island, Russia. Since yeast and bacteria coexist in a variety of habitats and can interact with each other, we analyzed the antibacterial activity of the collection of yeast strains towards E. coli cells using a fluorescent bacterial reporter. It was uncovered that the Sakhalin strains for the most part stimulate bacterial growth, while most of the strains found on the Kamchatka Peninsula possess inhibitory properties. Moreover, the samples with the most pronounced antibacterial activity, identified as members of the genus Cryptococcus (Naganishia), were found in a gorge in the vicinity of Pauzhetka village on the Kamchatka Peninsula on wormwood (Artemisia vulgaris) and thistle (Onopordum acanthium). Our data indicate that the combination of a plant and its growth site is important for the emergence of yeast strains capable of secreting antibacterial compounds.
Collapse
Affiliation(s)
- O. V. Shulenina
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, Gatchina, 188300 Russian Federation
| | - E. A. Sukhanova
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, Gatchina, 188300 Russian Federation
| | - B. F. Yarovoy
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, Gatchina, 188300 Russian Federation
| | - E. A. Tolstyko
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, Gatchina, 188300 Russian Federation
| | - A. L. Konevega
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, Gatchina, 188300 Russian Federation
- Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 195251 Russian Federation
- National Research Centre “Kurchatov Institute”, Moscow, 123182 Russian Federation
| | - A. Paleskava
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, Gatchina, 188300 Russian Federation
- Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 195251 Russian Federation
| |
Collapse
|
16
|
Cerqueira Melo RDC, Martins AA, Melo ALF, Vicente JCP, Sturaro MC, Arantes JP, Rossato L, de Souza GHDA, Simionatto S. Investigating the Antimicrobial Potential of 560 Compounds from the Pandemic Response Box and COVID Box against Resistant Gram-Negative Bacteria. Antibiotics (Basel) 2024; 13:723. [PMID: 39200023 PMCID: PMC11350835 DOI: 10.3390/antibiotics13080723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
Antimicrobial resistance (AMR) has emerged as a significant threat to public health, particularly in infections caused by critically important Gram-negative bacteria. The development of novel antibiotics has its limitations, and therefore it is crucial to explore alternative strategies to effectively combat infections with resistant pathogens. In this context, the present study investigated the antibacterial potency of 560 compounds against the multidrug-resistant (MDR) strains of Klebsiella pneumoniae and Serratia marcescens. The evaluated compounds were selected from the Pandemic Response Box (PRB) and COVID Box (CB) and subjected to assays to determine the inhibitory concentration (IC), minimum bactericidal concentration (MBC), and biofilm formation. Further, the effects of these compounds on membrane integrity were assessed through protein quantification. Several of the evaluated compounds, including fusidic acid, MMV1580853, and MMV1634399, exhibited a significant reduction in biofilm formation and growth in K. pneumoniae. Trimethoprim exhibited potential against S. marcescens. The IC values of the compounds indicated significant microbial growth inhibition at various concentrations. These findings underscore the potency of the existing antibiotics and novel compounds in combating the MDR strains of bacteria. The importance of reconsidering the known antibiotics and utilizing drug repositioning strategies to address the increasing risk of AMR is highlighted.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Simone Simionatto
- Health Sciences Research Laboratory, Federal University of Grande Dourados (UFGD), Dourados 79804970, Mato Grosso do Sul, Brazil; (R.d.C.C.M.)
| |
Collapse
|
17
|
Noukrati H, Hamdan Y, Marsan O, El Fatimy R, Cazalbou S, Rey C, Barroug A, Combes C. Sodium fusidate loaded apatitic calcium phosphates: Adsorption behavior, release kinetics, antibacterial efficacy, and cytotoxicity assessment. Int J Pharm 2024; 660:124331. [PMID: 38866083 DOI: 10.1016/j.ijpharm.2024.124331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
The present work reports the adsorption, release, antibacterial properties, and in vitro cytotoxicity of sodium fusidate (SF) associated with a carbonated calcium phosphate bone cement. The adsorption study of SF on cement powder compared to stoichiometric hydroxyapatite and nanocrystalline carbonated apatite was investigated to understand the interaction between this antibiotic and the calcium phosphate phases involved in the cement formulation and setting reaction. The adsorption data revealed a fast kinetic process. However, the evolution of the amount of adsorbed SF was well described by a Freundlich-type isotherm characterized by a low adsorption capacity of the materials toward the SF molecule. The in vitro release results indicated a prolonged and controlled SF release for up to 34 days. The SF amounts eluted daily were at a therapeutic level (0.5-2 mg/L) and close to the antibiotic minimum inhibitory concentration (0.1-0.9 mg/L). Furthermore, the release data fitting and modeling suggested that the drug release occurred mainly by a diffusion mechanism. The antibacterial activity showed the effectiveness of SF released from the formulated cements against Staphylococcus aureus. Furthermore, the biological in vitro study demonstrated that the tested cements didn't show any cytotoxicity towards human peripheral blood mononuclear cells and did not significantly induce inflammation markers like IL-8.
Collapse
Affiliation(s)
- Hassan Noukrati
- Cadi Ayyad University, Faculty of Sciences Semlalia (SCIMATOP), Bd Prince My Abdellah, BP 2390, 40000 Marrakech, Morocco; CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, ENSIACET, 4 Allée Emile Monso, 31030 Toulouse Cedex 4, France; Institute of Biological Sciences, ISSB, Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir 43150, Morocco.
| | - Yousra Hamdan
- Institute of Biological Sciences, ISSB, Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir 43150, Morocco
| | - Olivier Marsan
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, ENSIACET, 4 Allée Emile Monso, 31030 Toulouse Cedex 4, France
| | - Rachid El Fatimy
- Institute of Biological Sciences, ISSB, Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir 43150, Morocco
| | - Sophie Cazalbou
- CIRIMAT, Université Toulouse 3 Paul Sabatier, Toulouse INP, CNRS, Université de Toulouse, 118 Route de Narbonne, 31062 Toulouse Cedex 9, France
| | - Christian Rey
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, ENSIACET, 4 Allée Emile Monso, 31030 Toulouse Cedex 4, France
| | - Allal Barroug
- Cadi Ayyad University, Faculty of Sciences Semlalia (SCIMATOP), Bd Prince My Abdellah, BP 2390, 40000 Marrakech, Morocco; Institute of Biological Sciences, ISSB, Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir 43150, Morocco
| | - Christèle Combes
- CIRIMAT, Toulouse INP, Université Toulouse 3 Paul Sabatier, CNRS, Université de Toulouse, ENSIACET, 4 Allée Emile Monso, 31030 Toulouse Cedex 4, France
| |
Collapse
|
18
|
González-López A, Larsson DSD, Koripella RK, Cain BN, Chavez MG, Hergenrother PJ, Sanyal S, Selmer M. Structures of the Staphylococcus aureus ribosome inhibited by fusidic acid and fusidic acid cyclopentane. Sci Rep 2024; 14:14253. [PMID: 38902339 PMCID: PMC11190147 DOI: 10.1038/s41598-024-64868-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024] Open
Abstract
The antibiotic fusidic acid (FA) is used to treat Staphylococcus aureus infections. It inhibits protein synthesis by binding to elongation factor G (EF-G) and preventing its release from the ribosome after translocation. While FA, due to permeability issues, is only effective against gram-positive bacteria, the available structures of FA-inhibited complexes are from gram-negative model organisms. To fill this knowledge gap, we solved cryo-EM structures of the S. aureus ribosome in complex with mRNA, tRNA, EF-G and FA to 2.5 Å resolution and the corresponding complex structures with the recently developed FA derivative FA-cyclopentane (FA-CP) to 2.0 Å resolution. With both FA variants, the majority of the ribosomal particles are observed in chimeric state and only a minor population in post-translocational state. As expected, FA binds in a pocket between domains I, II and III of EF-G and the sarcin-ricin loop of 23S rRNA. FA-CP binds in an identical position, but its cyclopentane moiety provides additional contacts to EF-G and 23S rRNA, suggesting that its improved resistance profile towards mutations in EF-G is due to higher-affinity binding. These high-resolution structures reveal new details about the S. aureus ribosome, including confirmation of many rRNA modifications, and provide an optimal starting point for future structure-based drug discovery on an important clinical drug target.
Collapse
Affiliation(s)
- Adrián González-López
- Department of Cell and Molecular Biology, Uppsala University, BMC, P.O. Box 596, 75124, Uppsala, Sweden
| | - Daniel S D Larsson
- Department of Cell and Molecular Biology, Uppsala University, BMC, P.O. Box 596, 75124, Uppsala, Sweden
| | - Ravi Kiran Koripella
- Department of Cell and Molecular Biology, Uppsala University, BMC, P.O. Box 596, 75124, Uppsala, Sweden
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University, Atlanta, USA
| | - Brett N Cain
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Martin Garcia Chavez
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Suparna Sanyal
- Department of Cell and Molecular Biology, Uppsala University, BMC, P.O. Box 596, 75124, Uppsala, Sweden
| | - Maria Selmer
- Department of Cell and Molecular Biology, Uppsala University, BMC, P.O. Box 596, 75124, Uppsala, Sweden.
| |
Collapse
|
19
|
Cao D, Yuan X, Jiang X, Wu T, Xiang Y, Ji Z, Liu J, Dong X, Bi K, Tønjum T, Xu K, Zhang Y. Antimicrobial and Antibiofilm Effects of Bithionol against Mycobacterium abscessus. Antibiotics (Basel) 2024; 13:529. [PMID: 38927195 PMCID: PMC11200778 DOI: 10.3390/antibiotics13060529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/10/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Mycobacterium abscessus (M. abscessus) is a multidrug-resistant nontuberculous mycobacterium (NTM) that is responsible for a wide spectrum of infections in humans. The lack of effective bactericidal drugs and the formation of biofilm make its clinical treatment very difficult. The FDA-approved drug library containing 3048 marketed and pharmacopeial drugs or compounds was screened at 20 μM against M. abscessus type strain 19977 in 7H9 medium, and 62 hits with potential antimicrobial activity against M. abscessus were identified. Among them, bithionol, a clinically approved antiparasitic agent, showed excellent antibacterial activity and inhibited the growth of three different subtypes of M. abscessus from 0.625 μM to 2.5 μM. We confirmed the bactericidal activity of bithionol by the MBC/MIC ratio being ≤4 and the time-kill curve study and also electron microscopy study. Interestingly, it was found that at 128 μg/mL, bithionol could completely eliminate biofilms after 48h, demonstrating an outstanding antibiofilm capability compared to commonly used antibiotics. Additionally, bithionol could eliminate 99.9% of biofilm bacteria at 64 μg/mL, 99% at 32 μg/mL, and 90% at 16 μg/mL. Therefore, bithionol may be a potential candidate for the treatment of M. abscessus infections due to its significant antimicrobial and antibiofilm activities.
Collapse
Affiliation(s)
- Dan Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiuzhi Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Tiantian Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yanghui Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhongkang Ji
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiaying Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xu Dong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Kefan Bi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Tone Tønjum
- Department of Microbiology, University of Oslo, Oslo University Hospital, 0424 Oslo, Norway
| | - Kaijin Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ying Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250117, China
| |
Collapse
|
20
|
Abstract
Covering: up to August 2023Terpenoids, which are widely distributed in animals, plants, and microorganisms, are a large group of natural products with diverse structures and various biological activities. They have made great contributions to human health as therapeutic agents, such as the anti-cancer drug paclitaxel and anti-malarial agent artemisinin. Accordingly, the biosynthesis of this important class of natural products has been extensively studied, which generally involves two major steps: hydrocarbon skeleton construction by terpenoid cyclases and skeleton modification by tailoring enzymes. Additionally, fungi (Ascomycota and Basidiomycota) serve as an important source for the discovery of terpenoids. With the rapid development of sequencing technology and bioinformatics approaches, genome mining has emerged as one of the most effective strategies to discover novel terpenoids from fungi. To date, numerous terpenoid cyclases, including typical class I and class II terpenoid cyclases as well as emerging UbiA-type terpenoid cyclases, have been identified, together with a variety of tailoring enzymes, including cytochrome P450 enzymes, flavin-dependent monooxygenases, and acyltransferases. In this review, our aim is to comprehensively present all fungal terpenoid cyclases identified up to August 2023, with a focus on newly discovered terpenoid cyclases, especially the emerging UbiA-type terpenoid cyclases, and their related tailoring enzymes from 2015 to August 2023.
Collapse
Affiliation(s)
- Pan Luo
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education of China, Jinan University, Guangzhou 510632, China.
| | - Jia-Hua Huang
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education of China, Jinan University, Guangzhou 510632, China.
| | - Jian-Ming Lv
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education of China, Jinan University, Guangzhou 510632, China.
| | - Gao-Qian Wang
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education of China, Jinan University, Guangzhou 510632, China.
| | - Dan Hu
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education of China, Jinan University, Guangzhou 510632, China.
| | - Hao Gao
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education of China, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
21
|
Dyett BP, Sarkar S, Yu H, Strachan J, Drummond CJ, Conn CE. Overcoming Therapeutic Challenges of Antibiotic Delivery with Cubosome Lipid Nanocarriers. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38690584 DOI: 10.1021/acsami.4c00921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Low discovery rates for new antibiotics, commercial disincentives to invest, and inappropriate use of existing drugs have created a perfect storm of antimicrobial resistance (AMR). This "silent pandemic" of AMR looms as an immense, global threat to human health. In tandem, many potential novel drug candidates are not progressed due to elevated hydrophobicity, which may result in poor intracellular internalization and undesirable serum protein binding. With a reducing arsenal of effective antibiotics, enabling technology platforms that improve the outcome of treatments, such as repurposing existing bioactive agents, is a prospective option. Nanocarrier (NC) mediated drug delivery is one avenue for amplifying the therapeutic outcome. Here, the performance of several antibiotic classes encapsulated within the lipid-based cubosomes is examined. The findings demonstrate that encapsulation affords significant improvements in drug concentration:inhibition outcomes and assists in other therapeutic challenges associated with internalization, enzyme degradation, and protein binding. We emphasize that a currently sidelined compound, novobiocin, became active and revealed a significant increase in inhibition against the pathogenic Gram-negative strain, Pseudomonas aeruginosa. Encapsulation affords co-delivery of multiple bioactives as a strategy for mitigating failure of monotherapies and tackling resistance. The rationale in optimized drug selection and nanocarrier choice is examined by transport modeling which agrees with experimental inhibition results. The results demonstrate that lipid nanocarrier encapsulation may alleviate a range of challenges faced by antibiotic therapies and increase the range of antibiotics available to treat bacterial infections.
Collapse
Affiliation(s)
- Brendan P Dyett
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
| | - Sampa Sarkar
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
| | - Haitao Yu
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
| | - Jamie Strachan
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
| | - Calum J Drummond
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
| | - Charlotte E Conn
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
| |
Collapse
|
22
|
Fang N, Wu L, Duan S, Li J. The Structural and Molecular Mechanisms of Mycobacterium tuberculosis Translational Elongation Factor Proteins. Molecules 2024; 29:2058. [PMID: 38731549 PMCID: PMC11085428 DOI: 10.3390/molecules29092058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Targeting translation factor proteins holds promise for developing innovative anti-tuberculosis drugs. During protein translation, many factors cause ribosomes to stall at messenger RNA (mRNA). To maintain protein homeostasis, bacteria have evolved various ribosome rescue mechanisms, including the predominant trans-translation process, to release stalled ribosomes and remove aberrant mRNAs. The rescue systems require the participation of translation elongation factor proteins (EFs) and are essential for bacterial physiology and reproduction. However, they disappear during eukaryotic evolution, which makes the essential proteins and translation elongation factors promising antimicrobial drug targets. Here, we review the structural and molecular mechanisms of the translation elongation factors EF-Tu, EF-Ts, and EF-G, which play essential roles in the normal translation and ribosome rescue mechanisms of Mycobacterium tuberculosis (Mtb). We also briefly describe the structure-based, computer-assisted study of anti-tuberculosis drugs.
Collapse
Affiliation(s)
- Ning Fang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai 200438, China; (N.F.); (L.W.)
| | - Lingyun Wu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai 200438, China; (N.F.); (L.W.)
| | - Shuyan Duan
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai 200438, China; (N.F.); (L.W.)
- College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang 277160, China
| | - Jixi Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai 200438, China; (N.F.); (L.W.)
| |
Collapse
|
23
|
Elsewedy HS, Shehata TM, Genedy SM, Siddiq KM, Asiri BY, Alshammari RA, Bukhari SI, Kola-Mustapha AT, Ramadan HA, Soliman WE. Enhancing the Topical Antibacterial Activity of Fusidic Acid via Embedding into Cinnamon Oil Nano-Lipid Carrier. Gels 2024; 10:268. [PMID: 38667687 PMCID: PMC11049292 DOI: 10.3390/gels10040268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Presently, antimicrobial resistance is of great risk to remarkable improvements in health conditions and infection management. Resistance to various antibiotics has been considered a great obstacle in their usage, necessitating alternative strategies for enhancing the antibacterial effect. Combination therapy has been recognized as a considerable strategy that could improve the therapeutic influence of antibacterial agents. Therefore, the aim of this study was to combine the antibacterial action of compounds of natural origin like fusidic acid (FA) and cinnamon essential oil (CEO) for synergistic effects. A distinctive nanoemulsion (NE) was developed using cinnamon oil loaded with FA. Applying the Box-Behnken design (BBD) approach, one optimized formula was selected and integrated into a gel base to provide an FA-NE-hydrogel for optimal topical application. The FA-NE-hydrogel was examined physically, studied for in vitro release, and investigated for stability upon storage at different conditions, at room (25 °C) and refrigerator (4 °C) temperatures, for up to 3 months. Ultimately, the NE-hydrogel preparation was inspected for its antibacterial behavior using multidrug-resistant bacteria and checked by scanning electron microscopy. The FA-NE-hydrogel formulation demonstrated a pH (6.32), viscosity (12,680 cP), and spreadability (56.7 mm) that are acceptable for topical application. The in vitro release could be extended for 6 h, providing 52.0%. The formulation was stable under both test conditions for up to 3 months of storage. Finally, the FA-NE-hydrogel was found to inhibit the bacterial growth of not only Gram-positive but also Gram-negative bacteria. The inhibition was further elucidated by a scanning electron micrograph, indicating the efficiency of CEO in enhancing the antibacterial influence of FA when combined in an NE system.
Collapse
Affiliation(s)
- Heba S. Elsewedy
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Diriyah 13713, Saudi Arabia
| | - Tamer M. Shehata
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Alhofuf 36362, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Shaymaa M. Genedy
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Diriyah 13713, Saudi Arabia
| | - Khuzama M. Siddiq
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Diriyah 13713, Saudi Arabia
| | - Bushra Y. Asiri
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Diriyah 13713, Saudi Arabia
| | - Rehab A. Alshammari
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Diriyah 13713, Saudi Arabia
| | - Sarah I. Bukhari
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Adeola T. Kola-Mustapha
- Department of Pharmaceutical Sciences, College of Pharmacy, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin 240003, Nigeria
| | - Heba A. Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Mansoura 11152, Egypt
| | - Wafaa E. Soliman
- Department of Microbiology and Immunology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Mansoura 11152, Egypt
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Alhofuf 36362, Saudi Arabia
| |
Collapse
|
24
|
Seo HW, Wassano NS, Amir Rawa MS, Nickles GR, Damasio A, Keller NP. A Timeline of Biosynthetic Gene Cluster Discovery in Aspergillus fumigatus: From Characterization to Future Perspectives. J Fungi (Basel) 2024; 10:266. [PMID: 38667937 PMCID: PMC11051388 DOI: 10.3390/jof10040266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
In 1999, the first biosynthetic gene cluster (BGC), synthesizing the virulence factor DHN melanin, was characterized in Aspergillus fumigatus. Since then, 19 additional BGCs have been linked to specific secondary metabolites (SMs) in this species. Here, we provide a comprehensive timeline of A. fumigatus BGC discovery and find that initial advances centered around the commonly expressed SMs where chemical structure informed rationale identification of the producing BGC (e.g., gliotoxin, fumigaclavine, fumitremorgin, pseurotin A, helvolic acid, fumiquinazoline). Further advances followed the transcriptional profiling of a ΔlaeA mutant, which aided in the identification of endocrocin, fumagillin, hexadehydroastechrome, trypacidin, and fumisoquin BGCs. These SMs and their precursors are the commonly produced metabolites in most A. fumigatus studies. Characterization of other BGC/SM pairs required additional efforts, such as induction treatments, including co-culture with bacteria (fumicycline/neosartoricin, fumigermin) or growth under copper starvation (fumivaline, fumicicolin). Finally, four BGC/SM pairs were discovered via overexpression technologies, including the use of heterologous hosts (fumicycline/neosartoricin, fumihopaside, sphingofungin, and sartorypyrone). Initial analysis of the two most studied A. fumigatus isolates, Af293 and A1160, suggested that both harbored ca. 34-36 BGCs. However, an examination of 264 available genomes of A. fumigatus shows up to 20 additional BGCs, with some strains showing considerable variations in BGC number and composition. These new BGCs present a new frontier in the future of secondary metabolism characterization in this important species.
Collapse
Affiliation(s)
- Hye-Won Seo
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI 53706, USA; (H.-W.S.); (N.S.W.); (M.S.A.R.); (G.R.N.)
| | - Natalia S. Wassano
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI 53706, USA; (H.-W.S.); (N.S.W.); (M.S.A.R.); (G.R.N.)
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo 13083-970, Brazil;
| | - Mira Syahfriena Amir Rawa
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI 53706, USA; (H.-W.S.); (N.S.W.); (M.S.A.R.); (G.R.N.)
| | - Grant R. Nickles
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI 53706, USA; (H.-W.S.); (N.S.W.); (M.S.A.R.); (G.R.N.)
| | - André Damasio
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo 13083-970, Brazil;
| | - Nancy P. Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI 53706, USA; (H.-W.S.); (N.S.W.); (M.S.A.R.); (G.R.N.)
- Department of Plant Pathology, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
25
|
Toufanian S, Mohammed J, Winterhelt E, Lofts A, Dave R, Coombes BK, Hoare T. A Nanocomposite Dynamic Covalent Cross-Linked Hydrogel Loaded with Fusidic Acid for Treating Antibiotic-Resistant Infected Wounds. ACS APPLIED BIO MATERIALS 2024; 7:1947-1957. [PMID: 38394042 DOI: 10.1021/acsabm.3c01293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is associated with high levels of morbidity and is considered a difficult-to-treat infection, often requiring nonstandard treatment regimens and antibiotics. Since over 40% of the emerging antibiotic compounds have insufficient solubility that limits their bioavailability and thus efficacy through oral or intravenous administration, it is crucial that alternative drug delivery products be developed for wound care applications. Existing effective treatments for soft tissue MRSA infections, such as fusidic acid (FA), which is typically administered orally, could also benefit from alternative routes of administration to improve local efficacy and bioavailability while reducing the required therapeutic dose. Herein, we report an antimicrobial poly(oligoethylene glycol methacrylate) (POEGMA)-based composite hydrogel loaded with fusidic acid-encapsulating self-assembled polylactic acid-b-poly(oligo(ethylene glycol) methyl ether methacrylate) (PLA-POEGMA) nanoparticles for the treatment of MRSA-infected skin wounds. The inclusion of the self-assembled nanoparticles (380 nm diameter when loaded with fusidic acid) does not alter the favorable mechanical properties and stability of the hydrogel in the context of its use as a wound dressing, while fusidic acid (FA) can be released from the hydrogel over ∼10 h via a diffusion-controlled mechanism. The antimicrobial studies demonstrate a clear zone of inhibition in vitro and a 1-2 order of magnitude inhibition of bacterial growth in vivo in an MRSA-infected full-thickness excisional murine wound model even at very low antibiotic doses. Our approach thus can both circumvent challenges in the local delivery of hydrophobic antimicrobial compounds and directly deliver antimicrobials into the wound to effectively combat methicillin-resistant infections using a fraction of the drug dose required using other clinically relevant strategies.
Collapse
Affiliation(s)
- Samaneh Toufanian
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Jody Mohammed
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Erica Winterhelt
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, Canada
| | - Andrew Lofts
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L7, Canada
| | - Ridhdhi Dave
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Brian K Coombes
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
26
|
Chang YT, Lin CY, Chen CJ, Hwang E, Alshetaili A, Yu HP, Fang JY. Neutrophil-targeted combinatorial nanosystems for suppressing bacteremia-associated hyperinflammation and MRSA infection to improve survival rates. Acta Biomater 2024; 174:331-344. [PMID: 38061677 DOI: 10.1016/j.actbio.2023.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 01/02/2024]
Abstract
There is currently no specific and effective treatment for bacteremia-mediated sepsis. Hence, this study engineered a combinatorial nanosystem containing neutrophil-targeted roflumilast-loaded nanocarriers and non-targeted fusidic acid-loaded nanoparticles to enable the dual mitigation of bacteremia-associated inflammation and methicillin-resistant Staphylococcus aureus (MRSA) infection. The targeted nanoparticles were developed by conjugating anti-lymphocyte antigen 6 complex locus G6D (Ly6G) antibody fragment on the nanoparticulate surface. The particle size and zeta potential of the as-prepared nanosystem were about 200 nm and -25 mV, respectively. The antibody-conjugated nanoparticles showed a three-fold increase in neutrophil internalization compared to the unfunctionalized nanoparticles. As a selective phosphodiesterase (PDE) 4 inhibitor, the roflumilast in the nanocarriers largely inhibited cytokine/chemokine release from the activated neutrophils. The fusidic acid-loaded nanocarriers were vital to eliminate biofilm MRSA colony by 3 log units. The nanoparticles drastically decreased the intracellular bacterial count compared to the free antibiotic. The in vivo mouse bioimaging demonstrated prolonged retention of the nanosystem in the circulation with limited organ distribution and liver metabolism. In the mouse bacteremia model, the multifunctional nanosystem produced a 1‒2 log reduction of MRSA burden in peripheral organs and blood. The functionalized nanosystem arrested the cytokine/chemokine overexpression greater than the unfunctionalized nanocarriers and free drugs. The combinatory nanosystem also extended the median survival time from 50 to 103 h. No toxicity from the nanoformulation was found based on histology and serum biochemistry. Furthermore, our data proved that the active neutrophil targeting by the versatile nanosystem efficiently alleviated MRSA infection and organ dysfunction caused by bacteremia. STATEMENT OF SIGNIFICANCE: Bacteremia-mediated sepsis poses a significant challenge in clinical practice, as there is currently no specific and effective treatment available. In our study, we have developed a novel combinatorial nanosystem to address this issue. Our nanosystem consists of neutrophil-targeted roflumilast-loaded nanocarriers and non-targeted fusidic acid-loaded nanoparticles, enabling the simultaneous mitigation of bacteremia-associated inflammation and MRSA infection. Our nanosystem demonstrated the decreased neutrophil activation, effective inhibition of cytokine release, elimination of MRSA biofilm colonies, and reduced intracellular bacterial counts. In vivo experiments showed prolonged circulation, limited organ distribution, and increased survival rates in a mouse bacteremia model. Importantly, our nanosystem exhibited no toxicity based on comprehensive assessments.
Collapse
Affiliation(s)
- Yen-Tzu Chang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Cheng-Yu Lin
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Chih-Jung Chen
- Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Erica Hwang
- Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Abdullah Alshetaili
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Huang-Ping Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan; School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan.
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan; Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan.
| |
Collapse
|
27
|
Haddad N, Ajaz J, Mansour L, Kasemodel R, Jarvis J, Jarad J, Gorski H, Carr M. A Review of the Clinical Utilization of Oral Antibacterial Therapy in the Treatment of Bone Infections in Adults. Antibiotics (Basel) 2023; 13:4. [PMID: 38275315 PMCID: PMC10812599 DOI: 10.3390/antibiotics13010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
Chronic osteomyelitis in adults is managed with prolonged courses of intravenous antibiotics in conjunction with surgical debridement of necrotic bone. Over the past 40 years, there has been no paradigm shift in this approach, as randomized controlled trials of this standard of care compared to alternatives such as prolonged oral antibiotics are scarce. However, there have been many small trials, case reports, and review papers evaluating the effectiveness of oral treatment for chronic osteomyelitis. The oral route for infections requiring prolonged treatment is intuitively and practically more favorable due to several advantages, the most important of which is the avoidance of long-term IV antimicrobial therapy with its complications, inconvenience, and cost. In this paper, we review the literature evaluating oral antibiotic therapy in the management of chronic bone infections since 1975. The majority of osteomyelitis infections are caused by Staphylococcus aureus, hence we focus on its treatment using oral antibiotics; however, we also emphasize subpopulations of patients with diabetes, implanted hardware, and with less common bacterial organisms. The primary objective of this review is to promulgate clinical recommendations on the use of oral antibiotics in bone infections in the context of initial therapy, transition from intravenous therapy, and the role of chronic suppression. The secondary objective is to summarize current knowledge of the specific oral antimicrobial agents that are commonly utilized, together with a synopsis of the available literature pertaining to their pharmacokinetic/pharmacodynamic properties and duration of therapy in bone infection.
Collapse
Affiliation(s)
- Nicholas Haddad
- College of Medicine, Central Michigan University, 1632 Stone St., Saginaw, MI 48603, USA
- CMU Medical Education Partners, Internal Medicine Residency Program, Central Michigan University, 1000 Houghton Ave., Saginaw, MI 48602, USA; (J.A.); (R.K.); (J.J.)
| | - Jibran Ajaz
- CMU Medical Education Partners, Internal Medicine Residency Program, Central Michigan University, 1000 Houghton Ave., Saginaw, MI 48602, USA; (J.A.); (R.K.); (J.J.)
| | - Lina Mansour
- Covenant HealthCare, 1447 N. Harrison St., Saginaw, MI 48602, USA; (L.M.); (M.C.)
| | - Robert Kasemodel
- CMU Medical Education Partners, Internal Medicine Residency Program, Central Michigan University, 1000 Houghton Ave., Saginaw, MI 48602, USA; (J.A.); (R.K.); (J.J.)
| | - Jennifer Jarvis
- Ascension St. Mary’s Hospital, 800 S. Washington Ave., Saginaw, MI 48601, USA;
| | - John Jarad
- CMU Medical Education Partners, Internal Medicine Residency Program, Central Michigan University, 1000 Houghton Ave., Saginaw, MI 48602, USA; (J.A.); (R.K.); (J.J.)
| | - Haley Gorski
- McLaren Bay Region, 1900 Columbus Ave., Bay City, MI 48708, USA;
| | - Maddie Carr
- Covenant HealthCare, 1447 N. Harrison St., Saginaw, MI 48602, USA; (L.M.); (M.C.)
| |
Collapse
|
28
|
Unni R, Andreani NA, Vallier M, Heinzmann SS, Taubenheim J, Guggeis MA, Tran F, Vogler O, Künzel S, Hövener JB, Rosenstiel P, Kaleta C, Dempfle A, Unterweger D, Baines JF. Evolution of E. coli in a mouse model of inflammatory bowel disease leads to a disease-specific bacterial genotype and trade-offs with clinical relevance. Gut Microbes 2023; 15:2286675. [PMID: 38059748 PMCID: PMC10730162 DOI: 10.1080/19490976.2023.2286675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a persistent inflammatory condition that affects the gastrointestinal tract and presents significant challenges in its management and treatment. Despite the knowledge that within-host bacterial evolution occurs in the intestine, the disease has rarely been studied from an evolutionary perspective. In this study, we aimed to investigate the evolution of resident bacteria during intestinal inflammation and whether- and how disease-related bacterial genetic changes may present trade-offs with potential therapeutic importance. Here, we perform an in vivo evolution experiment of E. coli in a gnotobiotic mouse model of IBD, followed by multiomic analyses to identify disease-specific genetic and phenotypic changes in bacteria that evolved in an inflamed versus a non-inflamed control environment. Our results demonstrate distinct evolutionary changes in E. coli specific to inflammation, including a single nucleotide variant that independently reached high frequency in all inflamed mice. Using ex vivo fitness assays, we find that these changes are associated with a higher fitness in an inflamed environment compared to isolates derived from non-inflamed mice. Further, using large-scale phenotypic assays, we show that bacterial adaptation to inflammation results in clinically relevant phenotypes, which intriguingly include collateral sensitivity to antibiotics. Bacterial evolution in an inflamed gut yields specific genetic and phenotypic signatures. These results may serve as a basis for developing novel evolution-informed treatment approaches for patients with intestinal inflammation.
Collapse
Affiliation(s)
- Rahul Unni
- Section Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Nadia Andrea Andreani
- Section Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Marie Vallier
- Section Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Silke S. Heinzmann
- Research Unit Analytical BioGeoChemistry, Helmholtz Munich, Neuherberg, Germany
| | - Jan Taubenheim
- Research Group Medical Systems Biology, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Martina A. Guggeis
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Florian Tran
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Olga Vogler
- Section Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Sven Künzel
- Section Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Jan-Bernd Hövener
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Astrid Dempfle
- Institute of Medical Informatics and Statistics, Kiel University, Kiel, Germany
| | - Daniel Unterweger
- Section Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - John F. Baines
- Section Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| |
Collapse
|
29
|
Chen S, Zhou P, Wu C, Wang J, Zhou Y, Zhang J, Wang B, Zhao H, Rao L, Li M, Yu F, Lin C. Polymyxin B and fusidic acid, a novel potent synergistic combination against Klebsiella pneumoniae and Escherichia coli isolates with polymyxin B resistance. Front Microbiol 2023; 14:1220683. [PMID: 37886061 PMCID: PMC10598591 DOI: 10.3389/fmicb.2023.1220683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
The increasing prevalence of multidrug-resistant (MDR) Gram-negative bacteria and comparatively limited options of antibiotics pose a major threat to public health worldwide. Polymyxin B is the last resort against extensively resistant Gram-negative bacterial infections. However, a large number of Gram-negative bacteria exhibited high-level resistance to Polymyxin B, bringing challenges for antimicrobial chemotherapy. Combination therapies using polymyxins and other antibiotics are recommended to treat multidrug-resistant pathogens. In this study, we selected Gram-negative bacterial strains, including Klebsiella pneumoniae and Escherichia coli, to explore whether fusidic acid and polymyxin B have a synergistic killing effect. Through broth microdilution, we observed that minimum inhibitory concentrations (MICs) against polymyxin B in the isolates tested were significantly reduced by the addition of fusidic acid. Notably, chequerboard analysis indicated a synergistic effect between polymyxin B and fusidic acid. In addition, subsequent time-kill experiments showed that the combination of polymyxin B and fusidic acid was more effective than a single drug in killing bacteria. Finally, our investigation utilizing the murine model revealed a higher survival rate in the combination therapy group compared to the monotherapy group. Our research findings provide evidence of the synergistic effect between polymyxin B and fusidic acid. Fusidic acid was shown to increase the sensitivity of multi-drug resistant E. coli and K. pneumoniae to polymyxin B, thereby enhancing its bactericidal activity. This study provides new insights into a potential strategy for overcoming polymyxin B resistance, however, further investigations are required to evaluate their feasibility in real clinical settings.
Collapse
Affiliation(s)
- Shuying Chen
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peiyao Zhou
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunyang Wu
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Wang
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Zhou
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bingjie Wang
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huilin Zhao
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lulin Rao
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Meilan Li
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunchan Lin
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
30
|
Douglas EJ, Laabei M. Staph wars: the antibiotic pipeline strikes back. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001387. [PMID: 37656158 PMCID: PMC10569064 DOI: 10.1099/mic.0.001387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
Antibiotic chemotherapy is widely regarded as one of the most significant medical advancements in history. However, the continued misuse of antibiotics has contributed to the rapid rise of antimicrobial resistance (AMR) globally. Staphylococcus aureus, a major human pathogen, has become synonymous with multidrug resistance and is a leading antimicrobial-resistant pathogen causing significant morbidity and mortality worldwide. This review focuses on (1) the targets of current anti-staphylococcal antibiotics and the specific mechanisms that confirm resistance; (2) an in-depth analysis of recently licensed antibiotics approved for the treatment of S. aureus infections; and (3) an examination of the pre-clinical pipeline of anti-staphylococcal compounds. In addition, we examine the molecular mechanism of action of novel antimicrobials and derivatives of existing classes of antibiotics, collate data on the emergence of resistance to new compounds and provide an overview of key data from clinical trials evaluating anti-staphylococcal compounds. We present several successful cases in the development of alternative forms of existing antibiotics that have activity against multidrug-resistant S. aureus. Pre-clinical antimicrobials show promise, but more focus and funding are required to develop novel classes of compounds that can curtail the spread of and sustainably control antimicrobial-resistant S. aureus infections.
Collapse
Affiliation(s)
| | - Maisem Laabei
- Department of Life Sciences, University of Bath, Bath BA2 7AY, UK
| |
Collapse
|
31
|
Esposito C, Kamper M, Trentacoste J, Galvin S, Pfister H, Wang J. Advances in the Cystic Fibrosis Drug Development Pipeline. Life (Basel) 2023; 13:1835. [PMID: 37763239 PMCID: PMC10532558 DOI: 10.3390/life13091835] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Cystic fibrosis is a genetic disease that results in progressive multi-organ manifestations with predominance in the respiratory and gastrointestinal systems. The significant morbidity and mortality seen in the CF population has been the driving force urging the CF research community to further advance treatments to slow disease progression and, in turn, prolong life expectancy. Enormous strides in medical advancements have translated to improvement in quality of life, symptom burden, and survival; however, there is still no cure. This review discusses the most current mainstay treatments and anticipated therapeutics in the CF drug development pipeline within the mechanisms of mucociliary clearance, anti-inflammatory and anti-infective therapies, restoration of the cystic fibrosis transmembrane conductance regulator (CFTR) protein (also known as highly effective modulator therapy (HEMT)), and genetic therapies. Ribonucleic acid (RNA) therapy, gene transfer, and gene editing are being explored in the hopes of developing a treatment and potential cure for people with CF, particularly for those not responsive to HEMT.
Collapse
Affiliation(s)
- Christine Esposito
- Division of Pulmonary, Critical Care and Sleep Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York, NY 11042, USA; (M.K.); (J.W.)
| | - Martin Kamper
- Division of Pulmonary, Critical Care and Sleep Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York, NY 11042, USA; (M.K.); (J.W.)
| | - Jessica Trentacoste
- Division of Pulmonary, Critical Care and Sleep Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York, NY 11042, USA; (M.K.); (J.W.)
| | - Susan Galvin
- Division of Pediatric Pulmonology, The Steven and Alexandra Cohen Children’s Medical Center, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lake Success, New York, NY 11042, USA;
| | - Halie Pfister
- Manhasset Office of Clinical Research, The Feinstein Institutes for Medical Research, Lake Success, New York, NY 11042, USA;
| | - Janice Wang
- Division of Pulmonary, Critical Care and Sleep Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York, NY 11042, USA; (M.K.); (J.W.)
- Manhasset Office of Clinical Research, The Feinstein Institutes for Medical Research, Lake Success, New York, NY 11042, USA;
| |
Collapse
|
32
|
Sułkowska-Ziaja K, Trepa M, Olechowska-Jarząb A, Nowak P, Ziaja M, Kała K, Muszyńska B. Natural Compounds of Fungal Origin with Antimicrobial Activity-Potential Cosmetics Applications. Pharmaceuticals (Basel) 2023; 16:1200. [PMID: 37765008 PMCID: PMC10535449 DOI: 10.3390/ph16091200] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
The phenomenon of drug resistance in micro-organisms necessitates the search for new compounds capable of combating them. Fungi emerge as a promising source of such compounds as they produce a wide range of secondary metabolites with bacteriostatic or fungistatic activity. These compounds can serve as alternatives for commonly used antibiotics. Furthermore, fungi also accumulate compounds with antiviral activity. This review focuses on filamentous fungi and macrofungi as sources of antimicrobial compounds. The article describes both individual isolated compounds and extracts that exhibit antibacterial, antifungal, and antiviral activity. These compounds are produced by the fruiting bodies and mycelium, as well as the biomass of mycelial cultures. Additionally, this review characterizes the chemical compounds extracted from mushrooms used in the realm of cosmetology; specifically, their antimicrobial activity.
Collapse
Affiliation(s)
- Katarzyna Sułkowska-Ziaja
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Monika Trepa
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Aldona Olechowska-Jarząb
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland
- Department of Microbiology, University Hospital, ul. Jakubowskiego 2, 30-688 Kraków, Poland
| | - Paweł Nowak
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków, Poland
| | - Marek Ziaja
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7, 31-034 Kraków, Poland
| | - Katarzyna Kała
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Bożena Muszyńska
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
33
|
Raileanu M, Borlan R, Campu A, Janosi L, Turcu I, Focsan M, Bacalum M. No country for old antibiotics! Antimicrobial peptides (AMPs) as next-generation treatment for skin and soft tissue infection. Int J Pharm 2023:123169. [PMID: 37356506 DOI: 10.1016/j.ijpharm.2023.123169] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/01/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
In recent years, the unprecedented rise of bacterial antibiotic resistance together with the lack of adequate therapies have made the treatment of skin infections and chronic wounds challenging, urging the scientific community to focus on the development of new and more efficient treatment strategies. In this context, there is a growing interest in the use of natural molecules with antimicrobial features, capable of supporting wound healing i.e., antimicrobial peptides (AMPs), for the treatment of skin and soft tissue infections. In this review, we give a short overview of the bacterial skin infections as well as some of the classic treatments used for topical application. We then summarize the AMPs classes, stressing the importance of the appropriate selection of the peptides based on their characteristics and physicochemical properties in order to maximize the antibacterial efficacy of the therapeutic systems against multi-drug resistant pathogens. Additionally, the present paper provides a comprehensive and rigorous assessment of the latest clinical trials investigating the efficacy of AMPs in the treatment of skin and soft tissue infections, highlighting the relevant outcomes. Seeking to obtain novel and improved compounds with synergistic activity, while also decreasing some of the known side effects of AMPs, we present two employed strategies using AMPs: (i) AMPs-conjugated nanosystems for systemic and topical drug delivery systems and (ii) antibiotics-peptide conjugates as a strategy to overcome antibiotics resistance. Finally, an important property of some of the AMPs used in wound treatment is highlighted: their ability to help in wound healing by generally promoting cell proliferation and migration, and in some cases re-epithelialization and angiogenesis among others. Thus, as the pursuit of improvement is an ongoing effort, this work presents the advances made in the treatment of skin and soft tissue infections along with their advantages and limitations, while the still remaining challenges are addressed by providing future prospects and strategies to overcome them.
Collapse
Affiliation(s)
- Mina Raileanu
- Department of Life and Environmental Physics, Horia Hulubei National Institute for Physics and Nuclear Engineering, Reactorului 30, Măgurele 077125, Romania
| | - Raluca Borlan
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, Treboniu Laurian No. 42, 400271 Cluj-Napoca, Romania
| | - Andreea Campu
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, Treboniu Laurian No. 42, 400271 Cluj-Napoca, Romania
| | - Lorant Janosi
- Molecular and Biomolecular Physics Department, National Institute for Research and Development of Isotopic and Molecular Technologies, 65-103 Donath Street, 400293 Cluj-Napoca, Romania
| | - Ioan Turcu
- Molecular and Biomolecular Physics Department, National Institute for Research and Development of Isotopic and Molecular Technologies, 65-103 Donath Street, 400293 Cluj-Napoca, Romania
| | - Monica Focsan
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, Treboniu Laurian No. 42, 400271 Cluj-Napoca, Romania.
| | - Mihaela Bacalum
- Department of Life and Environmental Physics, Horia Hulubei National Institute for Physics and Nuclear Engineering, Reactorului 30, Măgurele 077125, Romania.
| |
Collapse
|
34
|
Shanbhag C, Saraogi I. Bacterial GTPases as druggable targets to tackle antimicrobial resistance. Bioorg Med Chem Lett 2023; 87:129276. [PMID: 37030567 DOI: 10.1016/j.bmcl.2023.129276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023]
Abstract
Small molecules as antibacterial agents have contributed immensely to the growth of modern medicine over the last several decades. However, the emergence of drug resistance among bacterial pathogens has undermined the effectiveness of the existing antibiotics. Thus, there is an exigency to address the antibiotic crisis by developing new antibacterial agents and identifying novel drug targets in bacteria. In this review, we summarize the importance of guanosine triphosphate hydrolyzing proteins (GTPases) as key agents for bacterial survival. We also discuss representative examples of small molecules that target bacterial GTPases as novel antibacterial agents, and highlight areas that are ripe for exploration. Given their vital roles in cell viability, virulence, and antibiotic resistance, bacterial GTPases are highly attractive antibacterial targets that will likely play a vital role in the fight against antimicrobial resistance.
Collapse
|
35
|
Lin LC, Chang SC, Ou YH, Liu TP, Lu JJ. Clonal Spreading of ST42 Staphylococcus haemolyticus Strains Occurs Possibly Due to fusB and tetK Resistant Genes and Capsule-Related Genes. Int J Mol Sci 2023; 24:ijms24076198. [PMID: 37047168 PMCID: PMC10094739 DOI: 10.3390/ijms24076198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Multi-drug resistant Staphylococcus haemolyticus is a frequent nosocomial invasive bacteremia pathogen in hospitals. Our previous analysis showed one of the predominant strains, ST42 originated from ST3, had only one multilocus sequence typing (MLST) variation among seven loci in SH1431; yet no significant differences in biofilm formation observed between ST42 and ST3, suggesting that other factors influence clonal lineage change. Whole genome sequencing was conducted on two isolates from ST42 and ST3 to find phenotypic and genotypic variations, and these variations were further validated in 140 clinical isolates. The fusidic acid- and tetracycline-resistant genes (fusB and tetK) were found only in CGMH-SH51 (ST42). Further investigation revealed consistent resistant genotypes in all isolates, with 46% and 70% of ST42 containing fusB and tetK, respectively. In contrast, only 23% and 4.2% ST3 contained these two genes, respectively. The phenotypic analysis also showed that ST42 isolates were highly resistant to fusidic acid (47%) and tetracycline (70%), compared with ST3 (23% and 4%, respectively). Along with drug-resistant genes, three capsule-related genes were found in higher percentage distributions in ST42 than in ST3 isolates. Our findings indicate that ST42 could become endemic in Taiwan, further constitutive surveillance is required to prevent the spread of this bacterium.
Collapse
|
36
|
Tatarusanu SM, Sava A, Profire BS, Pinteala T, Jitareanu A, Iacob AT, Lupascu F, Simionescu N, Rosca I, Profire L. New Smart Bioactive and Biomimetic Chitosan-Based Hydrogels for Wounds Care Management. Pharmaceutics 2023; 15:pharmaceutics15030975. [PMID: 36986836 PMCID: PMC10060009 DOI: 10.3390/pharmaceutics15030975] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Wound management represents a continuous challenge for health systems worldwide, considering the growing incidence of wound-related comorbidities, such as diabetes, high blood pressure, obesity, and autoimmune diseases. In this context, hydrogels are considered viable options since they mimic the skin structure and promote autolysis and growth factor synthesis. Unfortunately, hydrogels are associated with several drawbacks, such as low mechanical strength and the potential toxicity of byproducts released after crosslinking reactions. To overcome these aspects, in this study new smart chitosan (CS)-based hydrogels were developed, using oxidized chitosan (oxCS) and hyaluronic acid (oxHA) as nontoxic crosslinkers. Three active product ingredients (APIs) (fusidic acid, allantoin, and coenzyme Q10), with proven biological effects, were considered for inclusion in the 3D polymer matrix. Therefore, six API-CS-oxCS/oxHA hydrogels were obtained. The presence of dynamic imino bonds in the hydrogels' structure, which supports their self-healing and self-adapting properties, was confirmed by spectral methods. The hydrogels were characterized by SEM, swelling degree, pH, and the internal organization of the 3D matrix was studied by rheological behavior. Moreover, the cytotoxicity degree and the antimicrobial effects were also investigated. In conclusion, the developed API-CS-oxCS/oxHA hydrogels have real potential as smart materials in wound management, based on their self-healing and self-adapting properties, as well as on the benefits of APIs.
Collapse
Affiliation(s)
- Simona-Maria Tatarusanu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Grigore T. Popa" of Iasi, 16 Universitatii Street, 700115 Iași, Romania
- Research & Development Department, Antibiotice Company, 1 ValeaLupului Street, 707410 Iasi, Romania
| | - Alexandru Sava
- Department of Analytical Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Grigore T. Popa" of Iasi, 16 Universitatii Street, 700115 Iași, Romania
| | - Bianca-Stefania Profire
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy "Grigore T. Popa" of Iasi, 16 Universitatii Street, 700115 Iași, Romania
| | - Tudor Pinteala
- Department of Orthopedics and Traumatology, Faculty of Medicine, University of Medicine and Pharmacy "Grigore T. Popa" of Iasi, 16 Universitatii Street, 700115 Iași, Romania
| | - Alexandra Jitareanu
- Department of Toxicology, Faculty of Pharmacy, University of Medicine and Pharmacy "Grigore T. Popa" of Iasi, 16 Universitatii Street, 700115 Iași, Romania
| | - Andreea-Teodora Iacob
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Grigore T. Popa" of Iasi, 16 Universitatii Street, 700115 Iași, Romania
| | - Florentina Lupascu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Grigore T. Popa" of Iasi, 16 Universitatii Street, 700115 Iași, Romania
| | - Natalia Simionescu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, "PetruPoni" Institute of Macromolecular Chemistry, 41A GrigoreGhica-Voda Alley, 700487 Iasi, Romania
| | - Irina Rosca
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, "PetruPoni" Institute of Macromolecular Chemistry, 41A GrigoreGhica-Voda Alley, 700487 Iasi, Romania
| | - Lenuta Profire
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy "Grigore T. Popa" of Iasi, 16 Universitatii Street, 700115 Iași, Romania
| |
Collapse
|
37
|
Salimova EV, Mozgovoj OS, Efimova SS, Ostroumova OS, Parfenova LV. 3-Amino-Substituted Analogues of Fusidic Acid as Membrane-Active Antibacterial Compounds. MEMBRANES 2023; 13:309. [PMID: 36984696 PMCID: PMC10056636 DOI: 10.3390/membranes13030309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 06/18/2023]
Abstract
Fusidic acid (FA) is an antibiotic with high activity against Staphylococcus aureus; it has been used in clinical practice since the 1960s. However, the narrow antimicrobial spectrum of FA limits its application in the treatment of bacterial infections. In this regard, this work aims both at the study of the antimicrobial effect of a number of FA amines and at the identification of their potential biological targets. In this way, FA analogues containing aliphatic and aromatic amino groups and biogenic polyamine, spermine and spermidine, moieties at the C-3 atom, were synthesized (20 examples). Pyrazinecarboxamide-substituted analogues exhibit a high antibacterial activity against S. aureus (MRSA) with MIC ≤ 0.25 μg/mL. Spermine and spermidine derivatives, along with activity against S. aureus, also inhibit the growth and reproduction of Gram-negative bacteria Escherichia coli, Acinetobacter baumannii, and Pseudomonas aeruginosa, and have a high fungicidal effect against Candida albicans and Cryptococcus neoformans. The study of the membrane activity demonstrated that the spermidine- and spermine-containing compounds are able to immerse into membranes and disorder the lipidsleading to a detergent effect. Moreover, spermine-based compounds are also able to form ion-permeable pores in the lipid bilayers mimicking the bacterial membranes. Using molecular docking, inhibition of the protein synthesis elongation factor EF-G was proposed, and polyamine substituents were shown to make the greatest contribution to the stability of the complexes of fusidic acid derivatives with biological targets. This suggests that the antibacterial effect of the obtained compounds may be associated with both membrane activity and inhibition of the elongation factor EF-G.
Collapse
Affiliation(s)
- Elena V. Salimova
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141 Prospect Oktyabrya, 450075 Ufa, Russia
| | - Oleg S. Mozgovoj
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141 Prospect Oktyabrya, 450075 Ufa, Russia
| | - Svetlana S. Efimova
- Institute of Cytology of Russian Academy of Sciences, 4 Tikhoretsky Prospect, 194064 Saint Petersburg, Russia
| | - Olga S. Ostroumova
- Institute of Cytology of Russian Academy of Sciences, 4 Tikhoretsky Prospect, 194064 Saint Petersburg, Russia
| | - Lyudmila V. Parfenova
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141 Prospect Oktyabrya, 450075 Ufa, Russia
| |
Collapse
|
38
|
Kim JY, Jegal BS, Koh AR, Park KS, Kim JB, Bae J. Growth inhibition by fusidic acid in cervical, thyroid, and breast carcinoma cell lines. Obstet Gynecol Sci 2023; 66:100-106. [PMID: 36575560 PMCID: PMC10025870 DOI: 10.5468/ogs.22267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE We investigated the effects of fusidic acid (FA) on human cervical, thyroid, and breast carcinoma cell lines to determine the potential usefulness of FA in cancer treatment. METHODS Six cancer cell lines (cervical cancer: Caski, HeLa; thyroid cancer: 8505C, TPC1; and breast cancer: MCF-7, MDA-MB-231) were treated with FA. Furthermore the changes in cell growth, cell cycle duration, and extent of apoptosis were analyzed. RESULTS After FA treatment, the cancer cells showed a decrease in growth rate. In the cell death assay, the cell populations were similar in each cell type after treatment with FA, indicating that growth inhibition by FA was not related to the induction of apoptosis. FA induced cell cycle arrest at a dose that inhibited growth rate, which varied in different cell types. G0/G1 phase arrest occurs in breast cancer, S phase arrest in 8505C thyroid cancer, and G2/M phase arrest in cervical cancer. These results indicate that FA reduces growth rates by inducing cell cycle arrest. CONCLUSION FA treatment can interfere with cell proliferation by inducing cell cycle arrest in human cervical, thyroid, and breast carcinoma cell lines. Thus, FA can be useful in treating human cervical, thyroid, and breast carcinomas.
Collapse
Affiliation(s)
- Jin Young Kim
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul,
Korea
| | - Bo Seul Jegal
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul,
Korea
| | - A Ra Koh
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul,
Korea
| | - Kyoung Sik Park
- Department Departments of Surgery, Konkuk University School of Medicine, Seoul,
Korea
| | | | - Jaeman Bae
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul,
Korea
| |
Collapse
|
39
|
Ziroglu N, Koluman A, Kaleci B, Tanriverdi B, Tanriverdi G, Kural A, Bilgili MG. The antibiotics supplemented bone cement improved the masquelet's induced membrane in a rat femur critical size defect model. Injury 2023; 54:329-338. [PMID: 36334950 DOI: 10.1016/j.injury.2022.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/10/2022] [Accepted: 10/24/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Masquelet technique is a two-stage surgical procedure used in the treatment of critical-size bone defects (CSD). Adding antibiotics to polymethylmethacrylate (PMMA) is still questionable to create higher quality induced membrane (IM). The aim of the study was to evaluate the effects of three antibiotic-supplemented cement, fusidic acid, teicoplanin, and gentamicin, on osteogenesis and IM progression applied to rat femur CSD model by comparing histopathological, biochemical, and immunohistochemical findings. METHODS Twenty-eight male rats were divided into four groups control, gentamicin (G), teicoplanin (T), and fusidic acid (FA). A 10 mm CSD was created in rat femurs. In the postoperative 4th week, intracardiac blood samples were collected for biochemical analysis of bone alkaline phosphatase (BALP), osteocalcin (OC), and tumor necrosis factor-alpha (TNF-α) levels. IMs obtained in secondary operation were fixed and prepared for histopathological scoring of membrane progression and immunohistochemical evaluation of rat-specific Transforming Growth Factor-Beta (TGF-β), Runt-related Transcription Factor 2 (Runx2), and Vascular Endothelial Growth Factor (VEGF) expressions. RESULTS Levels of BALP and OC in serum didn't change among groups significantly while serum TNF-α levels significantly decreased in all antibiotic groups compared to the control group (P = 0.017). Histological scores of groups FA and T were significantly higher than those of groups Control and G (P = 0.0007). IMs of groups T and FA showed good progression while those of groups Control and G were also moderately progressed. A significant increase in TGF-β expression was observed in group G and FA (P = 0.001) while a significant increase in the expression of VEGF was observed in groups G and T compared to the control group (P = 0.036). CONCLUSIONS The bone cement impregnated with thermostable and safe antibiotics, gentamicin, fusidic acid, and teicoplanin can increase osteogenesis and support IM progression by increasing the expressions of TGF-β and VEGF. Anabolic effects of induced membranes used in the treatment of critical-size bone defects can be enhanced by antibiotic-supplemented PMMAs applied by altering the original technique.
Collapse
Affiliation(s)
- Nezih Ziroglu
- Department of Orthopedics and Traumatology, Acibadem Mehmet Ali Aydinlar University School of Medicine, Acibadem Atakent Hospital, Kucukcekmece/Istanbul, Turkey.
| | - Alican Koluman
- Department of Orthopedics and Traumatology, Istanbul Bakirkoy Dr. Sadi Konuk Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Belisa Kaleci
- Department of Histology and Embryology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Bulent Tanriverdi
- Department of Orthopedics and Traumatology, Istanbul Bakirkoy Dr. Sadi Konuk Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Gamze Tanriverdi
- Department of Histology and Embryology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Alev Kural
- Department of Biochemistry, Istanbul Bakirkoy Dr. Sadi Konuk Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Mustafa Gokhan Bilgili
- Department of Orthopedics and Traumatology, Istanbul Bakirkoy Dr. Sadi Konuk Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
40
|
Li XY, Lv JM, Cao ZQ, Wang GQ, Lin FL, Chen GD, Qin SY, Hu D, Gao H, Yao XS. Biosynthetic characterization of the antifungal fernane-type triterpenoid polytolypin for generation of new analogues via combinatorial biosynthesis. Org Biomol Chem 2023; 21:851-857. [PMID: 36602159 DOI: 10.1039/d2ob02158g] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Fernane-type triterpenoids are a small group of natural products mainly found in plants and fungi with a wide range of biological activities. Polytolypin is a representative fernane-type triterpenoid from fungi and possesses potent antifungal activity. So far, biosynthesis of fungal-derived fernane-type triterpenoids has not been characterized, which hinders the expansion of their structural diversity using biosynthetic approaches. Herein, we identified the biosynthetic gene cluster of polytolypin and elucidated its biosynthetic pathway through heterologous expression in Aspergillus oryzae NSAR1, which involves a new triterpene cyclase for the biosynthesis of the hydrocarbon skeleton motiol, followed by multiple oxidations via three P450 enzymes. Moreover, two new triterpene cyclases for the biosynthesis of two other fernane-type skeletons isomotiol and fernenol were identified from fungi, and were individually co-expressed with the three P450 enzymes involved in polytolypin biosynthesis. These studies led to the generation of 13 fernane-type triterpenoids including eight new compounds, and two of them showed stronger antifungal activity towards Candida albicans FIM709 than polytolypin.
Collapse
Affiliation(s)
- Xin-Yu Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China.
| | - Jian-Ming Lv
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China.
| | - Zhi-Qin Cao
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China.
| | - Gao-Qian Wang
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China.
| | - Fu-Long Lin
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China.
| | - Guo-Dong Chen
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China.
| | - Sheng-Ying Qin
- Clinical Experimental Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Dan Hu
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China. .,Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hao Gao
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China. .,Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China.
| | - Xin-Sheng Yao
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China. .,Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
41
|
Dallo M, Patel K, Hebert AA. Topical Antibiotic Treatment in Dermatology. Antibiotics (Basel) 2023; 12:antibiotics12020188. [PMID: 36830098 PMCID: PMC9952385 DOI: 10.3390/antibiotics12020188] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/18/2023] Open
Abstract
Many indications in dermatology can be effectively managed with topical antibiotics, including acne vulgaris, wound infections, secondarily infected dermatitis, and impetigo. Dermatologists must be familiar with the wide spectrum of topical antibiotics available, including indications, mechanisms of action, adverse events, and spectra of activity. Dermatologists must also keep antibiotic resistance in mind when utilizing these medications. Due to the widespread use of topical antibiotics and their importance in dermatology, a literature review was performed using a systematic search of PubMed and Google Scholar with the terms topical antibiotics, skin infections, dermatology, antimicrobials, and inflammatory dermatoses to identify English-language articles published between 1965-2022 from any country. Relevant publications were manually reviewed for additional content. The following literature review will summarize the common topical antibiotics used in dermatology.
Collapse
Affiliation(s)
- Matthew Dallo
- Department of Dermatology, The University of Texas Health Houston, Houston, TX 77030, USA
- Correspondence: (M.D.); (K.P.); Tel.: +1-713-500-8278 (M.D. & K.P.)
| | - Kavina Patel
- Department of Dermatology, The University of Texas Health Houston, Houston, TX 77030, USA
- Correspondence: (M.D.); (K.P.); Tel.: +1-713-500-8278 (M.D. & K.P.)
| | - Adelaide A. Hebert
- Department of Dermatology and Pediatrics, The University of Texas Health Houston, Houston, TX 77030, USA
| |
Collapse
|
42
|
Yadav M, Dhyani S, Joshi P, Awasthi S, Tanwar S, Gupta V, Rathore DK, Chaudhuri S. Formic acid, an organic acid food preservative, induces viable-but-non-culturable state, and triggers new Antimicrobial Resistance traits in Acinetobacter baumannii and Klebsiella pneumoniae. Front Microbiol 2022; 13:966207. [PMID: 36504816 PMCID: PMC9730046 DOI: 10.3389/fmicb.2022.966207] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
Numerous human pathogens, especially Gram-negative bacteria, are able to enter the viable-but-non-culturable (VBNC) state when they are exposed to environmental stressors and pose the risk of being resuscitated and causing infection after the removal of the trigger. Widely used food preservatives like weak organic acids are potential VBNC inducers in food processing and packaging facilities but have only been reported for food-borne pathogens. In the present study, it is demonstrated for the first time that one such agent, formic acid (FA), can induce a VBNC state at food processing, storage, and distribution temperatures (4, 25, and 37°C) with a varied time of treatment (days 4-10) in pathogenic Gram-negative bacteria Acinetobacter baumannii and Klebsiella pneumoniae. The use of hospital-associated pathogens is critical based on the earlier reports that demonstrated the presence of these bacteria in hospital kitchens and commonly consumed foods. VBNC induction was validated by multiple parameters, e.g., non-culturability, metabolic activity as energy production, respiratory markers, and membrane integrity. Furthermore, it was demonstrated that the removal of FA was able to resuscitate VBNC with an increased expression of multiple virulence and Antimicrobial Resistance (AMR) genes in both pathogens. Since food additives/preservatives are significantly used in most food manufacturing facilities supplying to hospitals, contamination of these packaged foods with pathogenic bacteria and the consequence of exposure to food additives emerge as pertinent issues for infection control, and control of antimicrobial resistance in the hospital setting.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Susmita Chaudhuri
- Department of Multidisciplinary Clinical and Translational Research, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
43
|
Conrado R, Gomes TC, Roque GSC, De Souza AO. Overview of Bioactive Fungal Secondary Metabolites: Cytotoxic and Antimicrobial Compounds. Antibiotics (Basel) 2022; 11:1604. [PMID: 36421247 PMCID: PMC9687038 DOI: 10.3390/antibiotics11111604] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 08/27/2023] Open
Abstract
Microorganisms are known as important sources of natural compounds that have been studied and applied for different purposes in distinct areas. Specifically, in the pharmaceutical area, fungi have been explored mainly as sources of antibiotics, antiviral, anti-inflammatory, enzyme inhibitors, hypercholesteremic, antineoplastic/antitumor, immunomodulators, and immunosuppressants agents. However, historically, the high demand for new antimicrobial and antitumor agents has not been sufficiently attended by the drug discovery process, highlighting the relevance of intensifying studies to reach sustainable employment of the huge world biodiversity, including the microorganisms. Therefore, this review describes the main approaches and tools applied in the search for bioactive secondary metabolites, as well as presents several examples of compounds produced by different fungi species with proven pharmacological effects and additional examples of fungal cytotoxic and antimicrobial molecules. The review does not cover all fungal secondary metabolites already described; however, it presents some reports that can be useful at any phase of the drug discovery process, mainly for pharmaceutical applications.
Collapse
Affiliation(s)
| | | | | | - Ana Olívia De Souza
- Development and Innovation Laboratory, Instituto Butantan, Avenida Vital Brasil, 1500, São Paulo 05503-900, SP, Brazil
| |
Collapse
|
44
|
Revin VV, Liyaskina EV, Parchaykina MV, Kuzmenko TP, Kurgaeva IV, Revin VD, Ullah MW. Bacterial Cellulose-Based Polymer Nanocomposites: A Review. Polymers (Basel) 2022; 14:4670. [PMID: 36365662 PMCID: PMC9654748 DOI: 10.3390/polym14214670] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/29/2022] [Accepted: 10/30/2022] [Indexed: 10/15/2023] Open
Abstract
Bacterial cellulose (BC) is currently one of the most popular environmentally friendly materials with unique structural and physicochemical properties for obtaining various functional materials for a wide range of applications. In this regard, the literature reporting on bacterial nanocellulose has increased exponentially in the past decade. Currently, extensive investigations aim at promoting the manufacturing of BC-based nanocomposites with other components such as nanoparticles, polymers, and biomolecules, and that will enable to develop of a wide range of materials with advanced and novel functionalities. However, the commercial production of such materials is limited by the high cost and low yield of BC, and the lack of highly efficient industrial production technologies as well. Therefore, the present review aimed at studying the current literature data in the field of highly efficient BC production for the purpose of its further usage to obtain polymer nanocomposites. The review highlights the progress in synthesizing BC-based nanocomposites and their applications in biomedical fields, such as wound healing, drug delivery, tissue engineering. Bacterial nanocellulose-based biosensors and adsorbents were introduced herein.
Collapse
Affiliation(s)
- Viktor V. Revin
- Department of Biotechnology, Biochemistry and Bioengineering, National Research Ogarev Mordovia State University, 430005 Saransk, Russia
| | - Elena V. Liyaskina
- Department of Biotechnology, Biochemistry and Bioengineering, National Research Ogarev Mordovia State University, 430005 Saransk, Russia
| | - Marina V. Parchaykina
- Department of Biotechnology, Biochemistry and Bioengineering, National Research Ogarev Mordovia State University, 430005 Saransk, Russia
| | - Tatyana P. Kuzmenko
- Department of Biotechnology, Biochemistry and Bioengineering, National Research Ogarev Mordovia State University, 430005 Saransk, Russia
| | - Irina V. Kurgaeva
- Department of Biotechnology, Biochemistry and Bioengineering, National Research Ogarev Mordovia State University, 430005 Saransk, Russia
| | - Vadim D. Revin
- Faculty of Architecture and Civil Engineering, National Research Ogarev Mordovia State University, 430005 Saransk, Russia
| | - Muhammad Wajid Ullah
- Biofuels Institute, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
45
|
Sun Y, Hong S, Chen H, Yin Y, Wang C. Production of Helvolic Acid in Metarhizium Contributes to Fungal Infection of Insects by Bacteriostatic Inhibition of the Host Cuticular Microbiomes. Microbiol Spectr 2022; 10:e0262022. [PMID: 36047778 PMCID: PMC9602595 DOI: 10.1128/spectrum.02620-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/15/2022] [Indexed: 01/04/2023] Open
Abstract
The nortriterpenoid helvolic acid (HA) has potent antibiotic activities and can be produced by different fungi, yet HA function remains elusive. Here, we report the chemical biology of HA production in the insect pathogen Metarhizium robertsii. After deletion of the core oxidosqualene cyclase gene in Metarhizium, insect survival rates were significantly increased compared to those of insects treated with the wild type and the gene-rescued strain during topical infections but not during injection assays to bypass insect cuticles. Further gnotobiotic infection of axenic Drosophila adults confirmed the HA contribution to fungal infection by inhibiting bacterial competitors in an inoculum-dependent manner. Loss of HA production substantially impaired fungal spore germination and membrane penetration abilities relative to the WT and gene-complemented strains during challenge with different Gram-positive bacteria. Quantitative microbiome analysis revealed that HA production could assist the fungus to suppress the Drosophila cuticular microbiomes by exerting a bacteriostatic rather than bactericidal effect. Our data unveil the chemical ecology of HA and highlight the fact that fungal pathogens have to cope with the host cuticular microbiomes prior to successful infection of hosts. IMPORTANCE Emerging evidence has shown that the plant and animal surface microbiomes can defend hosts against fungal parasite infections. The strategies employed by fungal pathogens to combat the antagonistic inhibition of insect surface bacteria are still elusive. In this study, we found that the potent antibiotic helvolic acid (HA) produced by the insect pathogen Metarhizium robertsii contributes to natural fungal infection of insect hosts. Antibiotic and gnotobiotic infection assays confirmed that HA could facilitate fungal infection of insects by suppression of the host cuticular microbiomes through its bacteriostatic instead of bactericidal activities. The data from this study provide insights into the novel chemical biology of fungal secondary metabolisms.
Collapse
Affiliation(s)
- Yanlei Sun
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Song Hong
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Haimin Chen
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Ying Yin
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Chengshu Wang
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
46
|
Khairullah AR, Sudjarwo SA, Effendi MH, Ramandinianto SC, Widodo A, Riwu KHP. A review of horses as a source of spreading livestock-associated methicillin-resistant Staphylococcus aureus to human health. Vet World 2022; 15:1906-1915. [PMID: 36313842 PMCID: PMC9615495 DOI: 10.14202/vetworld.2022.1906-1915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
Livestock-associated methicillin-resistant Staphylococcus aureus (LA-MRSA) was first discovered in horses in 1989. Since then, LA-MRSA has begun to be considered an important strain of pathogenic bacteria in horses, which can cause LA-MRSA infection and colonization in humans with public health impacts. The anterior nares are the primary site of LA-MRSA colonization in horses, although LA-MRSA colonization may also occur in the gastrointestinal tract in horses. LA-MRSA-infected horses typically exhibit clinical infection or may not exhibit clinical infection. There are two potential risks associated with LA-MRSA colonization in horses: The possibility of disease development in horses infected with LA-MRSA and the possibility of LA-MRSA transfer to humans and other horses. The diagnosis of LA-MRSA in horses can be made by conducting in vitro sensitivity testing for oxacillin and cefoxitin, and then followed by a molecular test using polymerase chain reaction. LA-MRSA transmission in animal hospitals and on farms is most likely due to contact with horses infected or colonized by LA-MRSA. The history of prior antibiotic administration, history of prior LA-MRSA colonization, and length of equine hospitalization were described as risk factors in cases of infection and colonization of LA-MRSA in horses. Nebulized antibiotics may be a viable alternative to use in horses, but nebulized antibiotics are only used in horses that are persistently colonized with LA-MRSA. Controlling the spread of LA-MRSA in horses can be done by regularly washing horses, eradicating vectors in horse stalls such as rats, and maintaining the cleanliness of the stable and animal hospital environment. Meanwhile, cleaning hands, using gloves, and donning protective clothes are ways that humans can prevent the transmission of LA-MRSA when handling horses. This review will explain the definition of LA-MRSA in general, LA-MRSA in horses, the epidemiology of LA-MRSA in horses, the diagnosis of LA-MRSA in horses, the transmission of LA-MRSA in horses, risk factors for spreading LA-MRSA in horses, public health impact, treatment of LA-MRSA infection in horses, and control of the spread of LA-MRSA in horses.
Collapse
Affiliation(s)
- Aswin Rafif Khairullah
- Doctoral Program in Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Jl. Mulyorejo, Surabaya, Jawa Timur 60115, Indonesia
| | - Sri Agus Sudjarwo
- Department of Veterinary Pharmacology, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Jl. Mulyorejo, Surabaya, Jawa Timur 60115, Indonesia
| | - Mustofa Helmi Effendi
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Jl. Mulyorejo, Surabaya, Jawa Timur 60115, Indonesia
| | | | - Agus Widodo
- Doctoral Program in Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Jl. Mulyorejo, Surabaya, Jawa Timur 60115, Indonesia
| | - Katty Hendriana Priscilia Riwu
- Doctoral Program in Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Kampus C Unair, Jl. Mulyorejo, Surabaya, Jawa Timur 60115, Indonesia
| |
Collapse
|
47
|
Rivera-Chávez J, Ceapă CD, Figueroa M. Biological Dark Matter Exploration using Data Mining for the Discovery of Antimicrobial Natural Products. PLANTA MEDICA 2022; 88:702-720. [PMID: 35697058 DOI: 10.1055/a-1795-0562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The discovery of novel antimicrobials has significantly slowed down over the last three decades. At the same time, humans rely increasingly on antimicrobials because of the progressive antimicrobial resistance in medical practices, human communities, and the environment. Data mining is currently considered a promising option in the discovery of new antibiotics. Some of the advantages of data mining are the ability to predict chemical structures from sequence data, anticipation of the presence of novel metabolites, the understanding of gene evolution, and the corroboration of data from multiple omics technologies. This review analyzes the state-of-the-art for data mining in the fields of bacteria, fungi, and plant genomic data, as well as metabologenomics. It also summarizes some of the most recent research accomplishments in the field, all pinpointing to innovation through uncovering and implementing the next generation of antimicrobials.
Collapse
Affiliation(s)
- José Rivera-Chávez
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Corina-Diana Ceapă
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mario Figueroa
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
48
|
Zou Y, Yan R, Wang H, Zhong K, Wang S. NIR‐Responsive Polyurethane Nanocomposites Based on PDA@FA Nanoparticles with Synergistic Antibacterial Effect. MACROMOL CHEM PHYS 2022. [DOI: 10.1002/macp.202200141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yuke Zou
- College of Biomass Science and Engineering Sichuan University Chengdu 610065 P. R. China
| | - Rui Yan
- College of Biomass Science and Engineering Sichuan University Chengdu 610065 P. R. China
- The Key Laboratory of Leather Chemistry and Engineering of Ministry of Education Sichuan University Chengdu 610065 P. R. China
| | - Haibo Wang
- College of Biomass Science and Engineering Sichuan University Chengdu 610065 P. R. China
- The Key Laboratory of Leather Chemistry and Engineering of Ministry of Education Sichuan University Chengdu 610065 P. R. China
| | - Kai Zhong
- College of Biomass Science and Engineering Sichuan University Chengdu 610065 P. R. China
| | - Shuang Wang
- College of Biomass Science and Engineering Sichuan University Chengdu 610065 P. R. China
- The Key Laboratory of Leather Chemistry and Engineering of Ministry of Education Sichuan University Chengdu 610065 P. R. China
| |
Collapse
|
49
|
Elsewedy HS, Shehata TM, Soliman WE. Shea Butter Potentiates the Anti-Bacterial Activity of Fusidic Acid Incorporated into Solid Lipid Nanoparticle. Polymers (Basel) 2022; 14:2436. [PMID: 35746012 PMCID: PMC9228747 DOI: 10.3390/polym14122436] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Fusidic acid (FA) is an efficient anti-bacterial drug proven to be efficient against a wide range of bacteria. Nevertheless, the main restriction in its formulation is the limited solubility. To avoid such an obstacle, the drug is incorporated into the lipid core of the nanolipid formulation. Consequently, the present study was an attempt to formulate nanolipid preparation, mainly, solid lipid nanoparticle (SLN) integrating FA. FA-SLN was prepared using shea butter as a lipid phase owing to its reported anti-bacterial activity. Different FA-SLNs were fabricated using the central composite design (CCD) approach. The optimized formula was selected and integrated into a hydrogel base to be efficiently used topically. FA-SLN-hydrogel was evaluated for its character, morphology, in vitro release and stability. The formula was examined for irritation reaction and finally evaluated for its anti-bacterial performance. The optimized formula showed particle size 283.83 nm and entrapment 73.057%. The formulated FA-SLN-hydrogel displayed pH 6.2, viscosity 15,610 cP, spreadability 51.1 mm and in vitro release 64.6% following 180 min. FA-SLN-hydrogel showed good stability for three months at different conditions (room temperature and refrigerator). It exhibited no irritation reaction on the treated rats. Eventually, shea butter displayed a noteworthy effect against bacterial growth that improved the effect of FA. This would indicate prospective anti-bacterial activity of FA when combined with shea butter in SLN formulation as a promising nanocarrier.
Collapse
Affiliation(s)
- Heba S. Elsewedy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Alhofuf 36362, Al-Ahsa, Saudi Arabia;
| | - Tamer M. Shehata
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Alhofuf 36362, Al-Ahsa, Saudi Arabia;
- Department of Pharmaceutics, College of Pharmacy, Zagazig University, Ash Sharqiyah, Zagazig 44519, Egypt
| | - Wafaa E. Soliman
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Alhofuf 36362, Al-Ahsa, Saudi Arabia;
- Department of Microbiology and Immunology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Mansoura 11152, Egypt
| |
Collapse
|
50
|
Wei X, Wang WG, Matsuda Y. Branching and converging pathways in fungal natural product biosynthesis. Fungal Biol Biotechnol 2022; 9:6. [PMID: 35255990 PMCID: PMC8902786 DOI: 10.1186/s40694-022-00135-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/19/2022] [Indexed: 12/15/2022] Open
Abstract
AbstractIn nature, organic molecules with great structural diversity and complexity are synthesized by utilizing a relatively small number of starting materials. A synthetic strategy adopted by nature is pathway branching, in which a common biosynthetic intermediate is transformed into different end products. A natural product can also be synthesized by the fusion of two or more precursors generated from separate metabolic pathways. This review article summarizes several representative branching and converging pathways in fungal natural product biosynthesis to illuminate how fungi are capable of synthesizing a diverse array of natural products.
Collapse
|