1
|
Lee JH, Song J, Hong S, Kim Y, Song M, Cho B, Wu T, Riley LW, Landegren U, Lee LP. Nanoplasmonic Rapid Antimicrobial-Resistance Point-of-Care Identification Device: RAPIDx. Adv Healthc Mater 2025; 14:e2402044. [PMID: 39205550 DOI: 10.1002/adhm.202402044] [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: 07/02/2024] [Revised: 08/05/2024] [Indexed: 09/04/2024]
Abstract
The emergence of antibiotic resistance has become a global health crisis, and everyone must arm themselves with wisdom to effectively combat the "silent tsunami" of infections that are no longer treatable with antibiotics. However, the overuse or inappropriate use of unnecessary antibiotics is still routine for administering them due to the unavailability of rapid, precise, and point-of-care assays. Here, a rapid antimicrobial-resistance point-of-care identification device (RAPIDx) is reported for the accurate and simultaneous identification of bacterial species (genotype) and target enzyme activity (phenotype). First, a contamination-free active target enzyme is extracted via the photothermal lysis of preconcentrated bacteria cells on a nanoplasmonic functional layer on-chip. Second, the rapid, precise identification of pathogens is achieved by the photonic rolling circle amplification of DNA on a chip. Third, the simultaneous identification of bacterial species (genotype) and target enzyme activity (phenotype) is demonstrated within a sample-to-answer 45 min operation via the RAPIDx. It is believed that the RAPIDx will be a valuable method for solving the bottleneck of employing on-chip nanotechnology for antibiotic-resistant bioassay and other infectious diseases.
Collapse
Affiliation(s)
- Jong-Hwan Lee
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, 94720, USA
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, 34114, South Korea
| | - Jihwan Song
- Department of Mechanical Engineering, Hanbat National University, Daejeon, 34158, South Korea
| | - SoonGweon Hong
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, 94720, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yun Kim
- Department of Mechanical Engineering, Hanbat National University, Daejeon, 34158, South Korea
| | - Minsun Song
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, 94720, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Byungrae Cho
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Tiffany Wu
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Lee W Riley
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Ulf Landegren
- Departments of Immunology, Genetics and Pathology, Uppsala University, Uppsala, SE-751 08, Sweden
| | - Luke P Lee
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, 94720, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Electrical Engineering and Computer Science, University of California Berkeley, Berkeley, CA, 94720, USA
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, South Korea
| |
Collapse
|
2
|
Shepperson OA, Harris PWR, Brimble MA, Cameron AJ. Thanatin and vinyl sulfide analogues as narrow spectrum antimicrobial peptides that synergise with polymyxin B. Front Pharmacol 2024; 15:1487338. [PMID: 39564120 PMCID: PMC11573584 DOI: 10.3389/fphar.2024.1487338] [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: 08/27/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
Thanatin is a β-hairpin antimicrobial peptide cyclised by a single disulfide bond that has shown potent broad-spectrum activity towards bacterial and fungal pathogens. Towards Gram-negative species, thanatin acts both by forming trans-membranal pores and inhibiting outer membrane biogenesis by binding to LptA and blocking lipopolysaccharide (LPS) transport. Inspired by previous modifications of thanatin, an analogue was prepared which demonstrated potent but selective activity towards E. coli. Furthermore, this compound was shown to act in synergy with the highly potent FDA-approved lipopeptide antibiotic polymyxin B, which engages LPS at the cytoplasmic membrane. Four analogues of thanatin in which the disulfide was substituted for vinyl sulfide bridge mimetics were prepared, all of which retained similar secondary structures. Two of these retained substantial potency and selectivity towards E. coli. Importantly, synergy with polymyxin B was also maintained for the lead analogue. The vinyl sulfide potentially offers a facile replacement strategy for labile disulfide bonds and the selective activity and drug synergy of the reported thanatin analogues is promising for the development of narrow spectrum antimicrobials with reduced likelihood of resistance emerging in clinical settings.
Collapse
Affiliation(s)
- Oscar A Shepperson
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Bio-Discovery, The University of Auckland, Auckland, New Zealand
| | - Paul W R Harris
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Bio-Discovery, The University of Auckland, Auckland, New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Bio-Discovery, The University of Auckland, Auckland, New Zealand
| | - Alan J Cameron
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Bio-Discovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
3
|
Alzard S, Exintaris B, Sarkar M, Grieve A, Chuang S, Coetzee R, Lim A. A global investigation into antimicrobial knowledge in medicine, pharmacy, nursing, dentistry and veterinary undergraduate students: A scoping review to inform future planetary health multidisciplinary education. BMC MEDICAL EDUCATION 2024; 24:1227. [PMID: 39468598 PMCID: PMC11520657 DOI: 10.1186/s12909-024-06253-w] [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: 06/26/2024] [Accepted: 10/24/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Inappropriate use of antimicrobials can push the environment out of balance, and cause unnecessary waste that can contaminate our soil, animals and waterways. Health professional education is committed to preparing students for antimicrobial stewardship (AMS) and supporting planetary health, but a more multidisciplinary action is needed to curb the expansion of antimicrobial resistance (AMR). The aim of this scoping review is to showcase the current antimicrobial knowledge of undergraduate students across the disciplines of medicine, pharmacy, nursing, dentistry and veterinary. This will consequently showcase the gaps and trends across the different disciplines and countries to help inform planetary health multidisciplinary undergraduate curriculums. METHODS A search of the existing literature published prior to December 2023 was conducted using CINAHL, EMBASE, MEDLINE, SCOPUS, and ERIC databases. Studies were excluded if they included postgraduate students or discussed the knowledge, attitude and practice (KAP) of students towards antimicrobial prescribing, AMR and AMS related to a specific learning activity. RESULTS A total of 144 articles were included. The most represented countries were India and Pakistan accounting for 17% and 8% of the studies, respectively. Single-disciplinary research accounted for approximately 80% of the studies. Medicine was the most represented discipline in both single-disciplinary and multidisciplinary research, appearing in approximately 62% of the studies, followed by pharmacy appearing in approximately 30% of the studies and dentistry appearing in approximately 18% of the studies. Three major priority themes were identified: students are more familiar with the term AMR compared to AMS; inappropriate use of antimicrobials is seen as the main driver of AMR; and the need for more training and education in the field of appropriate antimicrobial prescribing, AMR and AMS. CONCLUSION This review has highlighted that there is a need for more AMS interprofessional education (IPE) activities in all five disciplines, and especially within the disciplines of nursing, veterinary and dentistry, as shown by a lack of multidisciplinary research in this area. Most of the knowledge assessments have just touched the surface of AMS and focused on inappropriate antimicrobial use alone. Interdisciplinary planetary health education needs to go beyond these skills and broaden the understanding of other factors that can contribute to AMR such as inappropriate disposal, environmental contamination, monitoring and surveillance, one health, false allergies, and more importantly, how each health professional can contribute to a team.
Collapse
Affiliation(s)
- Shahd Alzard
- Monash University, Melbourne, VIC, 3052, Australia
| | | | | | | | - Sara Chuang
- Monash University, Melbourne, VIC, 3052, Australia
| | - Renier Coetzee
- School of Public Health, University of the Western Cape, Cape Town, South Africa
| | - Angelina Lim
- Monash University, Melbourne, VIC, 3052, Australia.
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, 3052, Australia.
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.
| |
Collapse
|
4
|
Lau CL, Neoh HM, Periyasamy P, Tg Abu Bakar Sidik TMI, Tan TL, Ramli R, Naina Mohamed I. Prevalence and clinical significance of the genotypic carriage among ESBL phenotype-negative Escherichia coli and Klebsiella pneumoniae clinical isolates in bacteremia: a study in a Malaysian tertiary center. Front Cell Infect Microbiol 2024; 14:1429830. [PMID: 39512590 PMCID: PMC11540778 DOI: 10.3389/fcimb.2024.1429830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024] Open
Abstract
Background Antimicrobial resistance (AMR) can lead to fatal consequences. AMR genes carriage by phenotypically susceptible bacteria, such as Extended-Spectrum β-Lactamases (ESBL)s in Enterobacteriaceae, have potential implications for AMR spread and therapeutic outcomes. This phenomenon should be investigated. Methods Positive blood cultures from hospitalized patients in a Malaysian tertiary center between April 2022 and March 2023 were reviewed. A total of 137 clinical isolates of Escherichia coli (E.coli), Klebsiella pneumoniae (K.pneumoniae), and Klebsiella oxytoca were included. The antibiotic susceptibility and ESBL phenotypes were determined by disk diffusion method and the identification of genotypes by multiplex polymerase chain reaction. The clinical characteristics and outcome information were extracted by reviewing patients' medical records to evaluate the clinical significance of the ESBL genotype-positive but phenotype-negative isolates in bacteremia. Results All 137 isolates were positive for at least one genotype (bla CTX-M, n = 71, 51.8%; bla SHV, n = 87, 63.5%; bla TEM, n = 95, 69.3%; bla OXA-1, n = 38, 27.7%). While bla CTX-M was proportionately higher in the ESBL phenotype-positive isolates than ESBL phenotype-negative isolates (33/37, 89.2% vs 38/100, 38%; p < 0.001), more than half of those harboring bla CTX-M remained susceptible to third-generation cephalosporins (3GC). The sensitivity (Sen) of bla CTX-M for ESBL phenotypes prediction was 89.19% (95% confidence interval [CI], 74.58 - 96.97%); however, specificity (Sp) was low (46.47%; 95% CI 39.75 - 53.32). The patient characteristics were similar among 98 ESBL phenotype-negative cases, except that the non-bla CTX-M carrier group had significantly more renal impairment (0/37 vs 7/61, p = 0.043) and gastrointestinal sources of bacteremia (9/37 vs 27/61, p = 0.047). No differences were observed in infection severity, in-hospital mortality, and length of stay (LOS) between the bla CTX-M and non-bla CTX-M carrier groups. Conclusion The current study provides insight into the gene carriage in E.coli and Klebsiella species clinical isolates, including bla CTX-M genotypes in antibiotic-susceptible strains from a Malaysian hospital. The ESBL encoding genotypes such as bla CTX-M presented substantially beyond one-third of the ESBL phenotype-negative or 3GC susceptible E.coli and K.pneumoniae isolated from bloodstream infection. Although clinical outcomes were not worsened with bla CTX-M genotype-positive but ESBL phenotype-negative isolates in bacteremia, the potential implications for AMR spread deserve further investigation.
Collapse
Affiliation(s)
- Chee Lan Lau
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
- Pharmacy Department, Hospital Canselor Tuanku Muhriz, Kuala Lumpur, Malaysia
| | - Hui-min Neoh
- UKM Medical Molecular Biology Institute (UMBI), National University of Malaysia, Kuala Lumpur, Malaysia
- Faculty of Health Sciences, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Petrick Periyasamy
- Medical Department, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Tg Mohd Ikhwan Tg Abu Bakar Sidik
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Toh Leong Tan
- Emergency Medicine Department, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Ramliza Ramli
- Department of Medical Microbiology and Immunology, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Isa Naina Mohamed
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
5
|
Campos DL, Canales CSC, Demarqui FM, Fernandes GFS, dos Santos CG, Prates JLB, da Silva IGM, Barros-Cordeiro KB, Báo SN, de Andrade LN, Abichabki N, Zacharias LV, de Campos MMA, dos Santos JL, Pavan FR. Screening of novel narrow-spectrum benzofuroxan derivatives for the treatment of multidrug-resistant tuberculosis through in silico, in vitro, and in vivo approaches. Front Microbiol 2024; 15:1487829. [PMID: 39464394 PMCID: PMC11502347 DOI: 10.3389/fmicb.2024.1487829] [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: 08/28/2024] [Accepted: 10/01/2024] [Indexed: 10/29/2024] Open
Abstract
Tuberculosis remains a serious global health threat, exacerbated by the rise of resistant strains. This study investigates the potential of two benzofuroxan (Bfx) derivatives, 5n and 5b, as targeted treatments for MDR-TB using in silico, in vitro, and in vivo methodologies. In vitro analyses showed that Bfx compounds have significant activity against Mtb H37Rv, with Bfx 5n standing out with a MIC90 of 0.09 ± 0.04 μM. Additionally, their efficacy against MDR and pre-XDR strains was superior compared to commercial drugs. These Bfx compounds have a narrow spectrum for mycobacteria, which helps avoid dysbiosis of the gut microbiota, and they also exhibit high selectivity and low toxicity. Synergism studies indicate that Bfx derivatives could be combined with rifampicin to enhance treatment efficacy and reduce its duration. Scanning electron microscopy revealed severe damage to the morphology of Mtb following treatment with Bfx 5n, showing significant distortions in the bacillary structures. Whole-genome sequencing of the 5n-resistant isolate suggests resistance mechanisms mediated by the Rv1855c gene, supported by in silico studies. In vivo studies showed that the 5n compound reduced the pulmonary load by 3.0 log10 CFU/mL, demonstrating superiority over rifampicin, which achieved a reduction of 1.23 log10 CFU/mL. In conclusion, Bfx derivatives, especially 5n, effectively address resistant infections caused by Mtb, suggesting they could be a solid foundation for future therapeutic developments against MDR-TB.
Collapse
Affiliation(s)
- Débora Leite Campos
- Tuberculosis Research Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| | - Christian Shleider Carnero Canales
- Tuberculosis Research Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
- School of Pharmacy, Biochemistry and Biotechnology, Santa Maria Catholic University, Arequipa, Peru
| | - Fernanda Manaia Demarqui
- Tuberculosis Research Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| | - Guilherme F. S. Fernandes
- Medicinal Chemistry Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
- School of Pharmacy, University College London, London, United Kingdom
| | - Camila Gonçalves dos Santos
- Tuberculosis Research Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| | - João Lucas B. Prates
- Medicinal Chemistry Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| | - Ingrid Gracielle Martins da Silva
- Microscopy and Microanalysis Laboratory, Cell Biology Department, Institute of Biological Sciences, University of Brasilia, Brasília, Brazil
| | - Karine Brenda Barros-Cordeiro
- Microscopy and Microanalysis Laboratory, Cell Biology Department, Institute of Biological Sciences, University of Brasilia, Brasília, Brazil
| | - Sônia Nair Báo
- Microscopy and Microanalysis Laboratory, Cell Biology Department, Institute of Biological Sciences, University of Brasilia, Brasília, Brazil
| | - Leonardo Neves de Andrade
- University of São Paulo – USPSchool of Pharmaceutical Sciences of Ribeirão Preto, , São Paulo, Brazil
| | - Nathália Abichabki
- University of São Paulo – USPSchool of Pharmaceutical Sciences of Ribeirão Preto, , São Paulo, Brazil
| | - Luísa Vieira Zacharias
- University of São Paulo – USPSchool of Pharmaceutical Sciences of Ribeirão Preto, , São Paulo, Brazil
| | - Marli Matiko Anraku de Campos
- Mycobacteriology Laboratory, Department of Clinical and Toxicological Analysis, Federal University of Santa Maria, Santa Maria, Brazil
| | - Jean Leandro dos Santos
- Medicinal Chemistry Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| | - Fernando Rogério Pavan
- Tuberculosis Research Laboratory, School of Pharmaceutical Sciences, São Paulo State University – UNESP, São Paulo, Brazil
| |
Collapse
|
6
|
Casado J, Olivan-Muro I, Algarate S, Chueca E, Salillas S, Velázquez-Campoy A, Piazuelo E, Fillat MF, Sancho J, Lanas Á, González A. Novel Drug-like HsrA Inhibitors Exhibit Potent Narrow-Spectrum Antimicrobial Activities against Helicobacter pylori. Int J Mol Sci 2024; 25:10175. [PMID: 39337660 PMCID: PMC11432330 DOI: 10.3390/ijms251810175] [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: 09/03/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Helicobacter pylori infection constitutes a silent pandemic of global concern. In the last decades, the alarming increase in multidrug resistance evolved by this pathogen has led to a marked drop in the eradication rates of traditional therapies worldwide. By using a high-throughput screening strategy, in combination with in vitro DNA binding assays and antibacterial activity testing, we identified a battery of novel drug-like HsrA inhibitors with MIC values ranging from 0.031 to 4 mg/L against several antibiotic-resistant strains of H. pylori, and minor effects against both Gram-negative and Gram-positive species of human microbiota. The most potent anti-H. pylori candidate demonstrated a high therapeutic index, an additive effect in combination with metronidazole and clarithromycin as well as a strong antimicrobial action against Campylobacter jejuni, another clinically relevant pathogen of phylum Campylobacterota. Transcriptomic analysis suggests that the in vivo inhibition of HsrA triggers lethal global disturbances in H. pylori physiology including the arrest of protein biosynthesis, malfunction of respiratory chain, detriment in ATP generation, and oxidative stress. The novel drug-like HsrA inhibitors described here constitute valuable candidates to a new family of narrow-spectrum antibiotics that allow overcoming the current resistome, protecting from dysbiosis, and increasing therapeutic options for novel personalized treatments against H. pylori.
Collapse
Affiliation(s)
- Javier Casado
- Group of Translational Research in Digestive Disease, Institute for Health Research Aragón (IIS Aragón), San Juan Bosco 13, 50009 Zaragoza, Spain
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Irene Olivan-Muro
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquilor (Edif. I+D), 50018 Zaragoza, Spain
| | - Sonia Algarate
- Microbiology Service, University Clinic Hospital Lozano Blesa, San Juan Bosco 15, 50009 Zaragoza, Spain
| | - Eduardo Chueca
- Group of Translational Research in Digestive Disease, Institute for Health Research Aragón (IIS Aragón), San Juan Bosco 13, 50009 Zaragoza, Spain
- Biomedical Research Networking Centre in Hepatic and Digestive Diseases (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Sandra Salillas
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquilor (Edif. I+D), 50018 Zaragoza, Spain
| | - Adrián Velázquez-Campoy
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquilor (Edif. I+D), 50018 Zaragoza, Spain
- Biomedical Research Networking Centre in Hepatic and Digestive Diseases (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Elena Piazuelo
- Group of Translational Research in Digestive Disease, Institute for Health Research Aragón (IIS Aragón), San Juan Bosco 13, 50009 Zaragoza, Spain
- Biomedical Research Networking Centre in Hepatic and Digestive Diseases (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Aragón Health Sciences Institute (IACS), San Juan Bosco 13, 50009 Zaragoza, Spain
| | - María F Fillat
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquilor (Edif. I+D), 50018 Zaragoza, Spain
| | - Javier Sancho
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquilor (Edif. I+D), 50018 Zaragoza, Spain
| | - Ángel Lanas
- Group of Translational Research in Digestive Disease, Institute for Health Research Aragón (IIS Aragón), San Juan Bosco 13, 50009 Zaragoza, Spain
- Biomedical Research Networking Centre in Hepatic and Digestive Diseases (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Department of Medicine, Psychiatry and Dermatology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Digestive Diseases Service, University Clinic Hospital Lozano Blesa, San Juan Bosco 15, 50009 Zaragoza, Spain
| | - Andrés González
- Group of Translational Research in Digestive Disease, Institute for Health Research Aragón (IIS Aragón), San Juan Bosco 13, 50009 Zaragoza, Spain
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquilor (Edif. I+D), 50018 Zaragoza, Spain
- Biomedical Research Networking Centre in Hepatic and Digestive Diseases (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| |
Collapse
|
7
|
Bassetti M, Cascio A, De Rosa FG, Meschiari M, Parrella R, Petrosillo N, Armuzzi A, Caprioli F, Dentali F, Pani M, Pilotto A, Restelli U, Sanguinetti M. Management of Clostridioides difficile infection: an Italian Delphi consensus. J Antimicrob Chemother 2024; 79:2103-2118. [PMID: 39008427 PMCID: PMC11368432 DOI: 10.1093/jac/dkae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Clostridioides difficile infection (CDI), a leading cause of nosocomial deaths, is a microbiota-mediated disease. As such, the use of broader spectrum antibiotics, such as vancomycin and metronidazole, can prime the gastrointestinal tract to become more prone to CDI recurrences. Fidaxomicin, a narrow-spectrum antibiotic, has been demonstrated to be superior in preventing recurrence and in preserving the intestinal microbiota; however, widespread employment worldwide has been hindered due to high acquisition costs. OBJECTIVES To integrate the currently available guidelines on the management of CDI and to shed light on the timeliest employment of fidaxomicin. METHODS An expert panel was gathered to obtain consensus using Delphi methodology on a series of statements regarding the management of CDI and on appropriate antibiotic use. RESULTS Consensus was reached on 21 of the 25 statements addressing the management of CDI. CONCLUSIONS Delphi methodology was used to achieve consensus on the management of CDI, on the identification of patients at risk of recurrences or severe infection, and on the most appropriate use of fidaxomicin, with the final aim of fostering clinical practice application of treatment algorithms proposed by previous guidelines, in absolute synergy. It could be an important tool to promote more appropriate and cost-effective CDI treatments in European settings with limited resources, like Italy.
Collapse
Affiliation(s)
- Matteo Bassetti
- Infectious Diseases Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Antonio Cascio
- Department PROMISE—Infectious and Tropical Diseases Unit, AOU Policlinico “P. Giaccone”, University of Palermo, 90127 Palermo, Italy
| | | | - Marianna Meschiari
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria di Modena, Policlinico di Modena, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Roberto Parrella
- Unit of Respiratory Infectious Diseases, Cotugno Hospital, Azienda Ospedaliera dei Colli, 80131 Naples, Italy
| | - Nicola Petrosillo
- Infection Prevention and Control Service, Fondazione Policlinico Universitario Campus Bio-Medico, 00127 Rome, Italy
| | - Alessandro Armuzzi
- IBD Unit, IRCCS Humanitas Research Hospital, Via A. Manzoni 56, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Flavio Caprioli
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20133 Milan, Italy
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico di Milano, 20122 Milan, Italy
| | - Francesco Dentali
- Division of Internal Medicine, Medical Center, Ospedale di Circolo & Fondazione Macchi, ASST Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Surgery, Insubria University, 21100 Varese, Italy
| | - Marcello Pani
- Hospital Pharmacy, Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Alberto Pilotto
- Department of Interdisciplinary Medicine, ‘Aldo Moro’ University of Bari, 70121 Bari, Italy
- Geriatrics Unit, Department of Geriatric Care, Neurology and Rehabilitation, Galliera Hospitals, 16128 Genova, Italy
| | | | - Maurizio Sanguinetti
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
8
|
Wang J, Gong R, Yang M, Wu X, Li Z, Huang H, Yan X, Wang D. A ruthenium single atom nanozyme-based antibiotic for the treatment of otitis media caused by Staphylococcus aureus. Front Chem 2024; 12:1439039. [PMID: 39263587 PMCID: PMC11387182 DOI: 10.3389/fchem.2024.1439039] [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: 05/27/2024] [Accepted: 08/14/2024] [Indexed: 09/13/2024] Open
Abstract
Staphylococcus aureus (S. aureus) infection is a primary cause of otitis media (OM), the most common disease for which children are prescribed antibiotics. However, the abuse of antibiotics has led to a global increase in antimicrobial resistance (AMR). Nanozymes, as promising alternatives to traditional antibiotics, are being extensively utilized to combat AMR. Here, we synthesize a series of single-atom nanozymes (metal-C3N4 SANzymes) by loading four metals (Ag, Fe, Cu, Ru) with antibacterial properties onto a crystalline g-C3N4. These metal-C3N4 display a rob-like morphology and well-dispersed metal atoms. Among them, Ru-C3N4 demonstrates the optimal peroxidase-like activity (285.3 U mg-1), comparable to that of horseradish peroxidase (267.7 U mg-1). In vitro antibacterial assays reveal that Ru-C3N4 significantly inhibits S. aureus growth compared with other metal-C3N4 even at a low concentration (0.06 mg mL-1). Notably, Ru-C3N4 acts as a narrow-spectrum nanoantibiotic with relative specificity against Gram-positive bacteria. Biofilms formed by S. aureus are easily degraded by Ru-C3N4 due to its high peroxidase-like activity. In vivo, Ru-C3N4 effectively eliminates S. aureus and relieves ear inflammation in OM mouse models. However, untreated OM mice eventually develop hearing impairment. Due to its low metal load, Ru-C3N4 does not exhibit significant toxicity to blood, liver, or kidney. In conclusion, this study presents a novel SANzyme-based antibiotic that can effectively eliminate S. aureus and treat S. aureus-induced OM.
Collapse
Affiliation(s)
- Jie Wang
- Nanozyme Synthesis Center, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Rui Gong
- Nanozyme Synthesis Center, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ming Yang
- Department of Otolaryngology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Xi Wu
- Nanozyme Synthesis Center, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ziwei Li
- Department of Clinical Laboratory, Shenshan Central Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Shanwei, China
| | - Haibing Huang
- Nanozyme Synthesis Center, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiyun Yan
- Nanozyme Synthesis Center, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, China
| | - Daji Wang
- Nanozyme Synthesis Center, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
9
|
Dehghan Manshadi M, Setoodeh P, Zare H. Systematic analysis of microorganisms' metabolism for selective targeting. Sci Rep 2024; 14:16446. [PMID: 39014020 PMCID: PMC11252421 DOI: 10.1038/s41598-024-65936-y] [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: 01/08/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024] Open
Abstract
Selective drugs with a relatively narrow spectrum can reduce the side effects of treatments compared to broad-spectrum antibiotics by specifically targeting the pathogens responsible for infection. Furthermore, combating an infectious pathogen, especially a drug-resistant microorganism, is more efficient by attacking multiple targets. Here, we combined synthetic lethality with selective drug targeting to identify multi-target and organism-specific potential drug candidates by systematically analyzing the genome-scale metabolic models of six different microorganisms. By considering microorganisms as targeted or conserved in groups ranging from one to six members, we designed 665 individual case studies. For each case, we identified single essential reactions as well as double, triple, and quadruple synthetic lethal reaction sets that are lethal for targeted microorganisms and neutral for conserved ones. As expected, the number of obtained solutions for each case depends on the genomic similarity between the studied microorganisms. Mapping the identified potential drug targets to their corresponding pathways highlighted the importance of key subsystems such as cell envelope biosynthesis, glycerophospholipid metabolism, membrane lipid metabolism, and the nucleotide salvage pathway. To assist in the validation and further investigation of our proposed potential drug targets, we introduced two sets of targets that can theoretically address a substantial portion of the 665 cases. We expect that the obtained solutions provide valuable insights into designing narrow-spectrum drugs that selectively cause system-wide damage only to the target microorganisms.
Collapse
Affiliation(s)
- Mehdi Dehghan Manshadi
- Department of Chemical Engineering, School of Chemical, Petroleum and Gas Engineering, Shiraz University, Shiraz, Iran
| | - Payam Setoodeh
- Department of Chemical Engineering, School of Chemical, Petroleum and Gas Engineering, Shiraz University, Shiraz, Iran.
- W Booth School of Engineering Practice and Technology, McMaster University, Hamilton, ON, Canada.
| | - Habil Zare
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA.
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
10
|
Wang Z, Cai W, Ning F, Sun W, Du J, Long S, Fan J, Chen X, Peng X. Dipicolylamine-Zn Induced Targeting and Photo-Eliminating of Pseudomonas aeruginosa and Drug-Resistance Gram-Positive Bacteria. Adv Healthc Mater 2024; 13:e2302490. [PMID: 37909241 DOI: 10.1002/adhm.202302490] [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: 08/25/2023] [Revised: 10/29/2023] [Indexed: 11/02/2023]
Abstract
The emergence of drug-resistant bacteria, particularly resistant strains of Gram-negative bacteria, such as Pseudomonas aeruginosa, poses a significant threat to public health. Although antibacterial photodynamic therapy (APDT) is a promising strategy for combating drug-resistant bacteria, actively targeted photosensitizers (PSs) remain unknown. In this study, a PS based on dipicolylamine (DPA), known as WZK-DPA-Zn, is designed for the selective identification of P. aeruginosa and drug-resistant Gram-positive bacteria. WZK-DPA-Zn exploits the synergistic effects of DPA-Zn2+ coordination and cellular uptake, which could effectively anchor P. aeruginosa within a brief period (10 min) without interference from other Gram-negative bacteria. Simultaneously, the cationic nature of WZK-DPA-Zn enhances its interaction with Gram-positive bacteria via electrostatic forces. Compared to traditional clinical antibiotics, WZK-DPA-Zn shows exceptional antibacterial activity without inducing drug resistance. This effectiveness is achieved using the APDT strategy when irradiated with white light or sunlight. The combination of WZK-DPA-Zn with Pluronic-based thermosensitive hydrogel dressings (WZK-DPA-Zn@Gel) effectively eliminates mixed bacterial infections and accelerates wound healing, thereby achieving a synergistic effect where "1+1>2." In summary, this study proposes a precise strategy employing DPA-Zn as the targeting moiety of a PS, facilitating the rapid elimination of P. aeruginosa and drug-resistant Gram-positive bacteria using APDT.
Collapse
Affiliation(s)
- Zuokai Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, P. R. China
| | - Wenlin Cai
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, P. R. China
| | - Fangrui Ning
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, P. R. China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, P. R. China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, P. R. China
| | - Saran Long
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, P. R. China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, P. R. China
| | - Xiaoqiang Chen
- State Key Laboratory of Fine Chemicals, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518071, P. R. China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, P. R. China
- State Key Laboratory of Fine Chemicals, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518071, P. R. China
| |
Collapse
|
11
|
Buendia P, Fernandez K, Raley C, Rahnavard A, Crandall KA, Castro JG. Hospital antimicrobial stewardship: profiling the oral microbiome after exposure to COVID-19 and antibiotics. Front Microbiol 2024; 15:1346762. [PMID: 38476940 PMCID: PMC10927822 DOI: 10.3389/fmicb.2024.1346762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/22/2024] [Indexed: 03/14/2024] Open
Abstract
Introduction During the COVID-19 Delta variant surge, the CLAIRE cross-sectional study sampled saliva from 120 hospitalized patients, 116 of whom had a positive COVID-19 PCR test. Patients received antibiotics upon admission due to possible secondary bacterial infections, with patients at risk of sepsis receiving broad-spectrum antibiotics (BSA). Methods The saliva samples were analyzed with shotgun DNA metagenomics and respiratory RNA virome sequencing. Medical records for the period of hospitalization were obtained for all patients. Once hospitalization outcomes were known, patients were classified based on their COVID-19 disease severity and the antibiotics they received. Results Our study reveals that BSA regimens differentially impacted the human salivary microbiome and disease progression. 12 patients died and all of them received BSA. Significant associations were found between the composition of the COVID-19 saliva microbiome and BSA use, between SARS-CoV-2 genome coverage and severity of disease. We also found significant associations between the non-bacterial microbiome and severity of disease, with Candida albicans detected most frequently in critical patients. For patients who did not receive BSA before saliva sampling, our study suggests Staphylococcus aureus as a potential risk factor for sepsis. Discussion Our results indicate that the course of the infection may be explained by both monitoring antibiotic treatment and profiling a patient's salivary microbiome, establishing a compelling link between microbiome and the specific antibiotic type and timing of treatment. This approach can aid with emergency room triage and inpatient management but also requires a better understanding of and access to narrow-spectrum agents that target pathogenic bacteria.
Collapse
Affiliation(s)
| | | | - Castle Raley
- The George Washington University Genomics Core, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Ali Rahnavard
- Department of Biostatistics and Bioinformatics, Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Keith A. Crandall
- The George Washington University Genomics Core, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
- Department of Biostatistics and Bioinformatics, Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Jose Guillermo Castro
- Division of Infectious Diseases, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
12
|
Nazli A, Tao W, You H, He X, He Y. Treatment of MRSA Infection: Where are We? Curr Med Chem 2024; 31:4425-4460. [PMID: 38310393 DOI: 10.2174/0109298673249381231130111352] [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: 02/19/2023] [Revised: 07/10/2023] [Accepted: 10/10/2023] [Indexed: 02/05/2024]
Abstract
Staphylococcus aureus is a leading cause of septicemia, endocarditis, pneumonia, skin and soft tissue infections, bone and joint infections, and hospital-acquired infections. In particular, methicillin-resistant Staphylococcus aureus (MRSA) is associated with high morbidity and mortality, and continues to be a major public health problem. The emergence of multidrug-resistant MRSA strains along with the wide consumption of antibiotics has made anti-MRSA treatment a huge challenge. Novel treatment strategies (e.g., novel antimicrobials and new administrations) against MRSA are urgently needed. In the past decade, pharmaceutical companies have invested more in the research and development (R&D) of new antimicrobials and strategies, spurred by favorable policies. All research articles were collected from authentic online databases, including Google Scholar, PubMed, Scopus, and Web of Science, by using different combinations of keywords, including 'anti-MRSA', 'antibiotic', 'antimicrobial', 'clinical trial', 'clinical phase', clinical studies', and 'pipeline'. The information extracted from articles was compared to information provided on the drug manufacturer's website and Clinical Trials.gov (https://clinicaltrials.gov/) to confirm the latest development phase of anti-MRSA agents. The present review focuses on the current development status of new anti-MRSA strategies concerning chemistry, pharmacological target(s), indications, route of administration, efficacy and safety, pharmacokinetics, and pharmacodynamics, and aims to discuss the challenges and opportunities in developing drugs for anti-MRSA infections.
Collapse
Affiliation(s)
- Adila Nazli
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Wenlan Tao
- Chongqing School, University of Chinese Academy of Sciences (UCAS Chongqing), Chongqing, 400714, China
| | - Hengyao You
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Xiaoli He
- Chongqing School, University of Chinese Academy of Sciences (UCAS Chongqing), Chongqing, 400714, China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| |
Collapse
|
13
|
Bergkessel M, Forte B, Gilbert IH. Small-Molecule Antibiotic Drug Development: Need and Challenges. ACS Infect Dis 2023; 9:2062-2071. [PMID: 37819866 PMCID: PMC10644355 DOI: 10.1021/acsinfecdis.3c00189] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Indexed: 10/13/2023]
Abstract
The need for new antibiotics is urgent. Antimicrobial resistance is rising, although currently, many more people die from drug-sensitive bacterial infections. The continued evolution of drug resistance is inevitable, fueled by pathogen population size and exposure to antibiotics. Additionally, opportunistic pathogens will always pose a threat to vulnerable patients whose immune systems cannot efficiently fight them even if they are sensitive to available antibiotics, according to clinical microbiology tests. These problems are intertwined and will worsen as human populations age, increase in density, and experience disruptions such as war, extreme weather events, or declines in standard of living. The development of appropriate drugs to treat all the world's bacterial infections should be a priority, and future success will likely require combinations of multiple approaches. However, the highest burden of bacterial infection is in Low- and Middle-Income Countries, where limited medical infrastructure is a major challenge. For effectively managing infections in these contexts, small-molecule-based treatments offer significant advantages. Unfortunately, support for ongoing small-molecule antibiotic discovery has recently suffered from significant challenges related both to the scientific difficulties in treating bacterial infections and to market barriers. Nevertheless, small-molecule antibiotics remain essential and irreplaceable tools for fighting infections, and efforts to develop novel and improved versions deserve ongoing investment. Here, we first describe the global historical context of antibiotic treatment and then highlight some of the challenges surrounding small-molecule development and potential solutions. Many of these challenges are likely to be common to all modalities of antibacterial treatment and should be addressed directly.
Collapse
Affiliation(s)
- Megan Bergkessel
- Division
of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, U.K.
| | - Barbara Forte
- Drug
Discovery Unit and Wellcome Centre for Anti-Infectives Research, Division
of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, U.K.
| | - Ian H. Gilbert
- Drug
Discovery Unit and Wellcome Centre for Anti-Infectives Research, Division
of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, U.K.
| |
Collapse
|
14
|
Flow-Based Fmoc-SPPS Preparation and SAR Study of Cathelicidin-PY Reveals Selective Antimicrobial Activity. Molecules 2023; 28:molecules28041993. [PMID: 36838983 PMCID: PMC9959817 DOI: 10.3390/molecules28041993] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
Antimicrobial peptides (AMPs) hold promise as novel therapeutics in the fight against multi-drug-resistant pathogens. Cathelicidin-PY (NH2-RKCNFLCKLKEKLRTVITSHIDKVLRPQG-COOH) is a 29-residue disulfide-cyclised antimicrobial peptide secreted as an innate host defence mechanism by the frog Paa yunnanensis (PY) and reported to possess broad-spectrum antibacterial and antifungal properties, exhibiting low cytotoxic and low hemolytic activity. Herein, we detail the total synthesis of cathelicidin-PY using an entirely on-resin synthesis, including assembly of the linear sequence by rapid flow Fmoc-SPPS and iodine-mediated disulfide bridge formation. By optimising a synthetic strategy to prepare cathelicidin-PY, this strategy was subsequently adapted to prepare a bicyclic head-to-tail cyclised derivative of cathelicidin-PY. The structure-activity relationship (SAR) of cathelicidin-PY with respect to the N-terminally positioned disulfide was further probed by preparing an alanine-substituted linear analogue and a series of lactam-bridged peptidomimetics implementing side chain to side chain cyclisation. The analogues were investigated for antimicrobial activity, secondary structure by circular dichroism (CD), and stability in human serum. Surprisingly, the disulfide bridge emerged as non-essential to antimicrobial activity and secondary structure but was amenable to synthetic modification. Furthermore, the synthetic AMP and multiple analogues demonstrated selective activity towards Gram-negative pathogen E. coli in physiologically relevant concentrations of divalent cations.
Collapse
|
15
|
Orlandi VT, Martegani E, Trivellin N, Bolognese F, Caruso E. Photo-Inactivation of Staphylococcus aureus by Diaryl-Porphyrins. Antibiotics (Basel) 2023; 12:antibiotics12020228. [PMID: 36830139 PMCID: PMC9951968 DOI: 10.3390/antibiotics12020228] [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: 12/28/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Photodynamic Antimicrobial Chemotherapy (PACT) has received great attention in recent years since it is an effective and promising modality for the treatment of human oral and skin infections with the advantage of bypassing pathogens' resistance to antimicrobials. Moreover, PACT applications demonstrated a certain activity in the inhibition and eradication of biofilms, overcoming the well-known tolerance of sessile communities to antimicrobial agents. In this study, 13 diaryl-porphyrins (mono-, di-cationic, and non-ionic) P1-P13 were investigated for their potential as photosensitizer anti-Staphylococcus aureus. The efficacy of the diaryl-porphyrins was evaluated through photo-inactivation tests. Crystal-violet staining combined with viable count techniques were aimed at assaying their anti-biofilm activity. Among the tested compounds, the neutral photosensitizer P4 was better than the cationic ones, irrespective of their corresponding binding rates. In particular, P4 was active in inhibiting the biofilm formation and in impairing the viability of the adherent and planktonic populations of a 24 h old biofilm. The inhibitory activity was also efficient against a methicillin resistant S. aureus strain. In conclusion, the diaryl-porphyrin family represents a reservoir of promising compounds for photodynamic applications against the pathogen S. aureus and in preventing the formation of biofilms that cause many infections to become chronic.
Collapse
Affiliation(s)
- Viviana Teresa Orlandi
- Department of Biotechnologies and Life Sciences, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
- Correspondence:
| | - Eleonora Martegani
- Department of Biotechnologies and Life Sciences, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| | - Nicola Trivellin
- Department of Industrial Engineering, University of Padova, Via Gradenigo 6A, 35131 Padova, Italy
| | - Fabrizio Bolognese
- Department of Biotechnologies and Life Sciences, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| | - Enrico Caruso
- Department of Biotechnologies and Life Sciences, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| |
Collapse
|
16
|
Synthesis and cytotoxic/antimicrobial screening of 2-alkenylimidazo[1,2-a]pyrimidines. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02997-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
17
|
Theuretzbacher U, Baraldi E, Ciabuschi F, Callegari S. Challenges and shortcomings of antibacterial discovery projects. Clin Microbiol Infect 2022; 29:610-615. [PMID: 36503116 PMCID: PMC10160915 DOI: 10.1016/j.cmi.2022.11.027] [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: 09/27/2022] [Revised: 11/18/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Antibacterial drug discovery activities are essential for filling clinical pipelines with promising clinical candidates. Little information is available about the challenges and shortcomings of small companies and academic institutions in performing these important discovery tasks. METHODS We performed a content analysis of 463 reviewer comments on 91 funding applications of antibacterial drug discovery projects submitted to two major global funders between 2016 and 2020 that had not proceeded further in the selection process. This quality assessment was complemented with the inputs (via e-mail) from a panel involving six antibiotic research and development (R&D) experts with long-standing expertise and experience in antibiotic drug discovery. RESULTS Common critical comments of reviewers are grouped into three main categories: scientific and technical shortcomings, unclear potential societal impact, and insufficient capability and expertise of the project team regarding the R&D process. Insufficient characterization of in vitro activity and/or testing of the hits/leads and insufficient antibacterial activity were the most common critical comments. Other areas of concern were insufficient or lack of differentiation from available drugs or projects with a long R&D history, and the research team's insufficient knowledge of a structured streamlined R&D process as reflected in severe gaps in the expertise of the R&D team. Little appreciation for the problem of the emergence of target-based resistance, especially in single-target approaches, and little awareness of toxicological issues, including approaches with historical liabilities were also commonly mentioned. The shortcomings identified through the analysis of funding applications are echoed by the results of the expert panel. DISCUSSION Our analysis identified an urgent need of strengthening the support for antibacterial drug discovery teams to help more projects reach such a quality to be eligible for global funders and private investors.
Collapse
Affiliation(s)
| | - Enrico Baraldi
- Department of Civil and Industrial Engineering, Uppsala University, Uppsala, Sweden
| | | | - Simone Callegari
- Department of Informatics and Media, Uppsala University, Uppsala, Sweden
| |
Collapse
|
18
|
Evolving antibiotic spectrum. Proc Natl Acad Sci U S A 2022; 119:e2214267119. [PMID: 36191212 PMCID: PMC9565054 DOI: 10.1073/pnas.2214267119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
19
|
Palmer JD, Foster KR. The evolution of spectrum in antibiotics and bacteriocins. Proc Natl Acad Sci U S A 2022; 119:e2205407119. [PMID: 36099299 PMCID: PMC9499554 DOI: 10.1073/pnas.2205407119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/05/2022] [Indexed: 12/02/2022] Open
Abstract
A key property of many antibiotics is that they will kill or inhibit a diverse range of microbial species. This broad-spectrum of activity has its evolutionary roots in ecological competition, whereby bacteria and other microbes use antibiotics to suppress other strains and species. However, many bacteria also use narrow-spectrum toxins, such as bacteriocins, that principally target conspecifics. Why has such a diversity in spectrum evolved? Here, we develop an evolutionary model to understand antimicrobial spectrum. Our first model recapitulates the intuition that broad-spectrum is best, because it enables a microbe to kill a wider diversity of competitors. However, this model neglects an important property of antimicrobials: They are commonly bound, sequestered, or degraded by the cells they target. Incorporating this toxin loss reveals a major advantage to narrow-spectrum toxins: They target the strongest ecological competitor and avoid being used up on less important species. Why then would broad-spectrum toxins ever evolve? Our model predicts that broad-spectrum toxins will be favored by natural selection if a strain is highly abundant and can overpower both its key competitor and other species. We test this prediction by compiling and analyzing a database of the regulation and spectrum of toxins used in inter-bacterial competition. This analysis reveals a strong association between broad-spectrum toxins and density-dependent regulation, indicating that they are indeed used when strains are abundant. Our work provides a rationale for why bacteria commonly evolve narrow-spectrum toxins such as bacteriocins and suggests that the evolution of antibiotics proper is a signature of ecological dominance.
Collapse
Affiliation(s)
- Jacob D. Palmer
- Department of Biology, University of Oxford, Oxford, OX1 3RB, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, United Kingdom
| | - Kevin R. Foster
- Department of Biology, University of Oxford, Oxford, OX1 3RB, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, United Kingdom
| |
Collapse
|
20
|
Zhao S, Wang Z, Lin Z, Wei G, Wen X, Li S, Yang X, Zhang Q, Jing C, Dai Y, Guo J, He Y. Drug Repurposing by Siderophore Conjugation: Synthesis and Biological Evaluation of Siderophore‐Methotrexate Conjugates as Antibiotics. Angew Chem Int Ed Engl 2022; 61:e202204139. [DOI: 10.1002/anie.202204139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Sheng Zhao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Zhi‐Peng Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
- Chongqing Institute of Green and Intelligent Technology Chinese Academy of Sciences Chongqing 400714 P. R. China
| | - Zihua Lin
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Guoxing Wei
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Xumei Wen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Siyu Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Xiaohong Yang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
- Chongqing Institute of Green and Intelligent Technology Chinese Academy of Sciences Chongqing 400714 P. R. China
| | - Qun Zhang
- Medicine Laboratory Children's Hospital of Chongqing Medical University Ministry of Education Key Laboratory of Child Development and Disorders 136 Zhongshan 2nd Rd Yuzhong, Chongqing 400014 P. R. China
| | - Chunmei Jing
- Medicine Laboratory Children's Hospital of Chongqing Medical University Ministry of Education Key Laboratory of Child Development and Disorders 136 Zhongshan 2nd Rd Yuzhong, Chongqing 400014 P. R. China
| | - Yuanwei Dai
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Jian Guo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| |
Collapse
|
21
|
Soybean peptide inhibits the biofilm of periodontopathic bacteria via bactericidal activity. Arch Oral Biol 2022; 142:105497. [PMID: 35849907 DOI: 10.1016/j.archoralbio.2022.105497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/09/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2022]
Abstract
OBJECTIVE This study aimed to clarify the antibacterial mechanism and antibiofilm effect of soybean-derived peptide BCBS-11 against periodontopathic bacteria. DESIGN The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of BCBS-11 against Porphyromonas gingivalis (P. gingivalis), Fusobacterium nucleatum (F. nucleatum), and Streptococcus mitis (S. mitis) were determined for the antibacterial mechanism. The effect of BCBS-11 on membrane permeability and depolarization activity were investigated using propidium iodide (PI) staining and 3, 3'-dipropylthiadicarbocyanine iodide (DiSC3-(5)) analysis. Monospecies and multispecies biofilms were cultured on 96-well plates. The amount of biofilm was determined using crystal violet staining to determine the inhibition of biofilm formation and the eradication of established biofilm using BCBS-11. The cytotoxicity of BCBS-11 was evaluated using 3-(4, 5-Dimethylthiazol-2-yl)- 2, 5-diphenyltetrazolium bromide (MTT) assay. RESULTS The MIC and MBC indicated the bactericidal activity of BCBS-11 against P. gingivalis and F. nucleatum. The PI staining revealed that BCBS-11 disrupted the bacterial membrane integrity. The DiSC3-(5) analysis indicated that BCBS-11 depolarized the bacterial cytoplasmic membrane. These results indicate the antimicrobial action of BCBS-11 through membrane disruption and the collapse of membrane electrochemical gradient. BCBS-11 significantly inhibited the monospecies biofilm formation of P. gingivalis and F. nucleatum and also inhibited dual-species biofilm. BCBS-11 was not cytotoxic toward human oral epithelial cells. CONCLUSIONS BCBS-11 inhibits the monospecies and multispecies biofilm formation of P. gingivalis and F. nucleatum, and their bactericidal activity results from membrane disruption.
Collapse
|
22
|
Zhao S, Wang ZP, Lin Z, Wei G, Wen X, Li S, Yang X, Zhang Q, Jing C, Dai Y, Guo J, He Y. Drug Repurposing by Siderophore Conjugation: Synthesis and Biological Evaluation of Siderophore‐Methotrexate Conjugates as Antibiotics. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202204139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Sheng Zhao
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Zhi-Peng Wang
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Zihua Lin
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Guoxing Wei
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Xumei Wen
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Siyu Li
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Xiaohong Yang
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Qun Zhang
- Chongqing Medical University Affiliated Children's Hospital Medicine Laboratory CHINA
| | - Chunmei Jing
- Chongqing Medical University Affiliated Children's Hospital Department of Clinical Laboratory CHINA
| | - Yuanwei Dai
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Jian Guo
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Yun He
- Chongqing University School of Pharmaceutical Sciences Daxuecheng South Road 401331 Chongqing CHINA
| |
Collapse
|
23
|
The Production of Antibiotics Must Be Reoriented: Repositioning Old Narrow-Spectrum Antibiotics, Developing New Microbiome-Sparing Antibiotics. Antibiotics (Basel) 2022; 11:antibiotics11070924. [PMID: 35884178 PMCID: PMC9311687 DOI: 10.3390/antibiotics11070924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 12/03/2022] Open
Abstract
The development of broad-spectrum antibiotics to control multidrug-resistant bacteria is an outdated business model. This strategy has led to the introduction of highly effective antibiotics, but their widespread use has contributed to the emergence of even broader antibiotic resistance. In a strategy to combat antimicrobial resistance, we believe that the use of narrow-spectrum antibiotics should be promoted. This should involve both the repositioning of old antibiotics and the reorientation of research and development towards new narrow-spectrum antibiotics with a low ecological impact. These antibiotics could be prescribed for common conditions such as sore throats and cystitis, which account for the bulk of antibiotic use in humans. Narrow-spectrum, targeted, microbiome-sparing antibiotics could help control antibiotic resistance while being economically sustainable. Their development and production should be supported by governments, which would ultimately benefit from reduced health care costs.
Collapse
|
24
|
Chou S, Zhang S, Guo H, Chang YF, Zhao W, Mou X. Targeted Antimicrobial Agents as Potential Tools for Modulating the Gut Microbiome. Front Microbiol 2022; 13:879207. [PMID: 35875544 PMCID: PMC9302920 DOI: 10.3389/fmicb.2022.879207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 06/13/2022] [Indexed: 12/11/2022] Open
Abstract
The gut microbiome plays a pivotal role in maintaining the health of the hosts; however, there is accumulating evidence that certain bacteria in the host, termed pathobionts, play roles in the progression of diseases. Although antibiotics can be used to eradicate unwanted bacteria, the side effects of antibiotic treatment lead to a great need for more targeted antimicrobial agents as tools to modulate the microbiome more precisely. Herein, we reviewed narrow-spectrum antibiotics naturally made by plants and microorganisms, followed by more targeted antibiotic agents including synthetic peptides, phage, and targeted drug delivery systems, from the perspective of using them as potential tools for modulating the gut microbiome for favorable effects on the health of the host. Given the emerging discoveries on pathobionts and the increasing knowledge on targeted antimicrobial agents reviewed in this article, we anticipate targeted antimicrobial agents will emerge as a new generation of a drug to treat microbiome-involved diseases.
Collapse
Affiliation(s)
- Shuli Chou
- Center for Infection and Immunity Studies, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Shiqing Zhang
- Center for Infection and Immunity Studies, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Huating Guo
- Center for Infection and Immunity Studies, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Yung-fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Wenjing Zhao
- Center for Infection and Immunity Studies, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Xiangyu Mou
- Center for Infection and Immunity Studies, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
25
|
Johnston CW, Badran AH. Natural and engineered precision antibiotics in the context of resistance. Curr Opin Chem Biol 2022; 69:102160. [PMID: 35660248 DOI: 10.1016/j.cbpa.2022.102160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022]
Abstract
Antibiotics are essential weapons in our fight against infectious disease, yet the consequences of broad-spectrum antibiotic use on microbiome stability and pathogen resistance are prompting investigations into more selective alternatives. Echoing the advent of precision medicine in oncology, precision antibiotics with focused activities are emerging as a means of addressing infections without damaging microbiomes or incentivizing resistance. Historically, antibiotic design principles have been gleaned from Nature, and reinvestigation of overlooked antibacterials is now providing scaffolds and targets for the design of pathogen-specific drugs. In this perspective, we summarize the biosynthetic and antibacterial mechanisms used to access these activities, and discuss how such strategies may be co-opted through engineering approaches to afford precision antibiotics.
Collapse
Affiliation(s)
- Chad W Johnston
- Department of Pharmacology & Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Ahmed H Badran
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
26
|
Precision Probiotics in Agroecosystems: Multiple Strategies of Native Soil Microbiotas for Conquering the Competitor Ralstonia solanacearum. mSystems 2022; 7:e0115921. [PMID: 35469423 PMCID: PMC9239239 DOI: 10.1128/msystems.01159-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ralstonia solanacearum (Rs), a soilborne phytopathogen, causes bacterial wilt disease in a broad range of hosts. Common approaches, for example, the direct reduction of the pathogen using classic single broad-spectrum probiotics, suffer from poor colonization efficiency, interference by resident microbiota, and nonnative-microorganism invasion. The soil microbiota plays an important role in plant health. Revealing the intrinsic linkage between the microbiome and the occurrence of disease and then applying it to agroecosystems for the precise control of soilborne diseases should be an effective strategy. Here, we surveyed the differences in the microbiome between healthy and diseased soils used for tomato planting across six climatic regions in China by using 16S rRNA amplicon and metagenomic sequencing. The roles of species associated with disease symptoms were further validated. Healthy soil possessed more diverse bacterial communities and more potential plant probiotics than diseased soil. Healthy soil simultaneously presented multiple strategies, including specifically antagonizing Rs, decreasing the gene expression of the type III secretion system of Rs, and competing for nutrition with Rs. Bacteria enriched in diseased samples promoted the progression of tomato bacterial wilt by strengthening the chemotaxis of pathogens. Therefore, Rs and its collaborators should be jointly combatted for disease suppression. Our research provides integrated insights into a multifaceted strategy for the biocontrol of tomato bacterial wilt based on the individual network of local microbiota. IMPORTANCE In the current work, the relationship between the soil microbiota and tomato bacterial wilt on a large scale offered us a comprehensive understanding of the disease. The delicate strategy of the microbiota in soil used for growing tomatoes to conquer the strong competitor, Rs, was revealed by microbiome research. The collaborators of Rs that coexist in a common niche with Rs strengthened our understanding of the pathogenesis of bacterial wilt. Bacteria enriched in healthy soil that antagonized pathogens with high specificity provide a novel view for ecofriendly probiotics mining. Our study offers new perspectives on soilborne-pathogen biocontrol in agroecosystems by decoding the rule of the natural ecosystem.
Collapse
|
27
|
Li H, Li T, Zhang L, Hu Q, Liao X, Jiang Q, Qiu X, Li L, Draheim RR, Huang Q, Zhou R. Antimicrobial compounds from an FDA-approved drug library with activity against Streptococcus suis. J Appl Microbiol 2021; 132:1877-1886. [PMID: 34800069 DOI: 10.1111/jam.15377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/12/2021] [Accepted: 11/17/2021] [Indexed: 02/03/2023]
Abstract
AIM Antimicrobial resistance (AMR) has become a global concern. Developing novel antimicrobials is one of the most effective approaches in tackling AMR. Considering its relatively low cost and risk, drug repurposing has been proposed as a valuable approach for novel antimicrobial discovery. The aim of this study was to screen for antimicrobial compounds against Streptococcus suis, an important zoonotic bacterial pathogen, from an Food and Drug Administration (FDA)-approved drug library. METHODS AND RESULTS In this study, we tested the antimicrobial activity of 1815 FDA-approved drugs against S. suis. Sixty-seven hits were obtained that showed a growth inhibition of more than 98%. After excluding already known antibiotics and antiseptics, 12 compounds were subjected to minimal inhibition concentration (MIC) assessment against S. suis. This showed that pralatrexate, daunorubicin (hydrochloride), teniposide, aclacinomycin A hydrochloride and floxuridine gave a relatively low MIC, ranging from 0.85 to 5.25 μg/ml. Apart from pralatrexate, the remaining four drugs could also inhibit the growth of antimicrobial-resistant S. suis. It was also demonstrated that these four drugs had better efficacy against Gram-positive bacteria than Gram-negative bacteria. Cytotoxicity assays showed that floxuridine and teniposide had a relatively high 50% cytotoxic concentration (CC50 ). The in vivo efficacy of floxuridine was analysed using a Galleria mellonella larvae infection model, and the results showed that floxuridine was effective in treating S. suis infection in vivo. CONCLUSIONS Five compounds from the FDA-approved drug library showed high antimicrobial activity against S. suis, among which floxuridine displayed potent in vivo efficacy that is worth further development. SIGNIFICANCE AND IMPACT OF STUDY Our study identified several antimicrobial compounds that are effective against S. suis, which provides a valuable starting point for further antimicrobial development.
Collapse
Affiliation(s)
- Haotian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tingting Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liangsheng Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qiao Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xia Liao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qinggen Jiang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiuxiu Qiu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lu Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China.,International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
| | - Roger R Draheim
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Qi Huang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China.,International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China.,International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
| |
Collapse
|
28
|
Duffy EM, Buurman ET, Chiang SL, Cohen NR, Uria-Nickelsen M, Alm RA. The CARB-X Portfolio of Nontraditional Antibacterial Products. ACS Infect Dis 2021; 7:2043-2049. [PMID: 34346202 DOI: 10.1021/acsinfecdis.1c00331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The growing prevalence of antibiotic-resistant bacterial pathogens and the lack of new medicines to treat the infections they cause remain a significant global threat. In recent years, this ongoing unmet need has encouraged more research groups to focus on the discovery and development of nontraditional antibacterial agents, ranging from anti-virulence strategies to bacteriophage and ways to modulate the microbiome. The Combating Antibiotic-Resistant Bacteria Biopharmaceutical Accelerator (CARB-X) is a global nonprofit public-private partnership dedicated to accelerating antibacterial-related research. Importantly, the CARB-X portfolio supports a wide variety of novel and innovative nontraditional programs to help the global antibacterial research ecosystem understand the potential that these modalities can play in the management or prevention of serious infections. We describe here the breadth of the CARB-X pipeline of novel nontraditional products.
Collapse
Affiliation(s)
- Erin M. Duffy
- CARB-X, Boston University, Boston, Massachusetts 02215, United States
| | - Ed T. Buurman
- CARB-X, Boston University, Boston, Massachusetts 02215, United States
| | - Su L. Chiang
- CARB-X, Boston University, Boston, Massachusetts 02215, United States
| | - Nadia R. Cohen
- CARB-X, Boston University, Boston, Massachusetts 02215, United States
| | | | - Richard A. Alm
- CARB-X, Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|
29
|
Altarac D, Gutch M, Mueller J, Ronsheim M, Tommasi R, Perros M. Challenges and opportunities in the discovery, development, and commercialization of pathogen-targeted antibiotics. Drug Discov Today 2021; 26:2084-2089. [PMID: 33610472 DOI: 10.1016/j.drudis.2021.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/25/2021] [Accepted: 02/12/2021] [Indexed: 11/27/2022]
Abstract
The use of antibiotics directly correlates with the increase in antimicrobial resistance (AMR). Targeting novel antibiotics to patients with multidrug-resistant (MDR) pathogens should enhance their durability and slow development of resistance. The discovery, development, and clinical adoption of pathogen-targeted antibiotics have been hampered by technical and regulatory challenges. Growing insights into bacterial physiology and mechanisms of resistance, innovative clinical trial designs, streamlined regulatory approval pathways, and availability of rapid bacterial diagnostics are recent developments that can help address those challenges. Pathogen-targeted antibiotics provide an opportunity to treat patients with the right drug at the right time, leading to improved patient outcomes and better antimicrobial stewardship. Patient-centered pricing and reimbursement reform is needed to incentivize innovation.
Collapse
Affiliation(s)
- David Altarac
- Entasis Therapeutics. Correspondence to Manos Perros, Entasis Therapeutics, 35 Gatehouse Drive, Waltham MA 02451, USA
| | - Michael Gutch
- Entasis Therapeutics. Correspondence to Manos Perros, Entasis Therapeutics, 35 Gatehouse Drive, Waltham MA 02451, USA
| | - John Mueller
- Entasis Therapeutics. Correspondence to Manos Perros, Entasis Therapeutics, 35 Gatehouse Drive, Waltham MA 02451, USA
| | - Matthew Ronsheim
- Entasis Therapeutics. Correspondence to Manos Perros, Entasis Therapeutics, 35 Gatehouse Drive, Waltham MA 02451, USA
| | - Ruben Tommasi
- Entasis Therapeutics. Correspondence to Manos Perros, Entasis Therapeutics, 35 Gatehouse Drive, Waltham MA 02451, USA
| | - Manos Perros
- Entasis Therapeutics. Correspondence to Manos Perros, Entasis Therapeutics, 35 Gatehouse Drive, Waltham MA 02451, USA.
| |
Collapse
|