1
|
Ge M, Jiang F, Lin H. Nanocatalytic medicine enabled next-generation therapeutics for bacterial infections. Mater Today Bio 2024; 29:101255. [PMID: 39381264 PMCID: PMC11459013 DOI: 10.1016/j.mtbio.2024.101255] [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] [Received: 07/01/2024] [Revised: 09/08/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024] Open
Abstract
The rapid rise of antibiotic-resistant strains and the persistence of biofilm-associated infections have significantly challenged global public health. Unfortunately, current clinical high-dose antibiotic regimens and combination therapies often fail to completely eradicate these infections, which can lead to adverse side effects and further drug resistance. Amidst this challenge, however, the burgeoning development in nanotechnology and nanomaterials brings hopes. This review provides a comprehensive summary of recent advancements in nanomaterials for treating bacterial infections. Firstly, the research progress of catalytic therapies in the field of antimicrobials is comprehensively discussed. Thereafter, we systematically discuss the strategies of nanomaterials for anti-bacterial infection therapies, including endogenous response catalytic therapy, exogenous stimulation catalytic therapy, and catalytic immunotherapy, in order to elucidate the mechanism of nanocatalytic anti-infections. Based on the current state of the art, we conclude with insights on the remaining challenges and future prospects in this rapidly emerging field.
Collapse
Affiliation(s)
- Min Ge
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Feng Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Han Lin
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, China
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200331, China
| |
Collapse
|
2
|
Zhang K, Yang N, Mao R, Hao Y, Teng D, Wang J. An amphipathic peptide combats multidrug-resistant Staphylococcus aureus and biofilms. Commun Biol 2024; 7:1582. [PMID: 39604611 PMCID: PMC11603143 DOI: 10.1038/s42003-024-07216-z] [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: 06/24/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
The emergence of drug-resistant Staphylococcus aureus (S. aureus) has resulted in infections in humans and animals that may lead to a crisis in the absence of highly effective drugs. Consequently, the development of alternative or complementary antimicrobial agents is urgently needed. Here, a series of peptides derived from AP138 were designed with high expression, antimicrobial activity, and antibiofilm properties via bioinformatics. Among them, the best derived peptide, A24 (S9A), demonstrated the greatest stability and bactericidal efficiency against multidrug-resistant S. aureus in a physiological environment, with a high hydrophobicity of 35%. This peptide exhibited superior performance compared to the preclinical or clinical antimicrobial peptides (AMPs). A24 displayed increased biocompatibility in vitro and in vivo, exhibiting a low hemolysis rate (less than 3%), minimal cytotoxicity (survival rate exceeding 85%), and no histotoxicity. A24 had the capacity to destroy cell walls, increase cell membrane permeability, and induce increases in intracellular ATP and ROS levels, which resulted in the rapid death of S. aureus. A24 inhibited the formation of early biofilms and eliminated both mature biofilms (40-50%) and persisters (99.9%). Therapeutic doses of A24 were shown to exhibit favorable safety profiles and bactericidal efficacy in vivo and could reduce bacterial loads of multidrug-resistant S. aureus by 4-5 log10 CFU/0.1g levels in mouse peritonitis and endometritis models. Furthermore, A24 increased the survival rate to 100% and exhibited anti-inflammatory properties in a mouse model. The aforementioned data illustrate the potential of A24 as a pharmaceutical agent for the treatment of bacterial infections, including peritonitis and endometritis, in animal husbandry with multidrug-resistant S. aureus infections.
Collapse
Affiliation(s)
- Kun Zhang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, 100081, Beijing, PR China
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, 100081, Beijing, PR China
| | - Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, 100081, Beijing, PR China
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, 100081, Beijing, PR China
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China.
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China.
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, 100081, Beijing, PR China.
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China.
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, 100081, Beijing, PR China.
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, 100081, Beijing, PR China.
| |
Collapse
|
3
|
Shi J, Xiao Y, Shen L, Wan C, Wang B, Zhou P, Zhang J, Han W, Yu F. Phenotypic and genomic analysis of the hypervirulent methicillin-resistant Staphylococcus aureus ST630 clone in China. mSystems 2024; 9:e0066424. [PMID: 39158330 PMCID: PMC11406941 DOI: 10.1128/msystems.00664-24] [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: 05/13/2024] [Accepted: 07/11/2024] [Indexed: 08/20/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) sequence type 630 (ST630) is a rarely reported lineage worldwide. This study aimed to trace the dissemination of the emerging MRSA ST630 clones in China and investigate their virulence potential. We collected 22 ST630-MRSA isolates from across China and performed whole-genome sequencing analysis and virulence characterization on these isolates. Epidemiological results showed that MRSA ST630 isolates were primarily isolated from pus/wound secretions, mainly originating from Jiangxi province, and carried diverse virulence and drug resistance genes. Staphylococcal cassette chromosome mec type V (SCCmec V) predominated (11/22, 50.0%) among the MRSA ST630 isolates. Interestingly, nearly half (45.5%) of the 22 ST630-MRSA isolates tested lacked intact SCCmec elements. Phylogenetic analysis demonstrated that ST630-MRSA could be divided into two distinct clades, with widespread dissemination mainly in Chinese regions. Five representative isolates were selected for phenotypic assays, including hemolysin activity, real-time fluorescence quantitative PCR, western blot analysis, hydrogen peroxide killing assay, blood killing assay, cell adhesion and invasion assay, and mouse skin abscess model. The results showed that, compared to the USA300-LAC strain, ST630 isolates exhibited particularly strong invasiveness and virulence in the aforementioned phenotypic assays. This study described the emergence of a highly virulent ST630-MRSA lineage and improved our insight into the molecular epidemiology of ST630 clones in China.IMPORTANCEMethicillin-resistant Staphylococcus aureus (MRSA) sequence type 630 (ST630) is an emerging clone with an increasing isolation rate in China. This study raises awareness of the hypervirulent MRSA ST630 clones in China and alerts people to their widespread dissemination. ST630-staphylococcal cassette chromosome mec V is a noteworthy clone in China, and we present the first comprehensive genetic and phenotypic analysis of this lineage. Our findings provide valuable insights for the prevention and control of infections caused by this emerging MRSA clone.
Collapse
Affiliation(s)
- Junhong Shi
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cailing Wan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peiyao Zhou
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weihua Han
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
4
|
Liu X, Xiong Y, Peng R, Zhang Y, Cai S, Deng Q, Yu Z, Wen Z, Chen Z, Hou T. Antibacterial activity and mechanisms of D-3263 against Staphylococcus aureus. BMC Microbiol 2024; 24:224. [PMID: 38926818 PMCID: PMC11201875 DOI: 10.1186/s12866-024-03377-3] [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: 02/19/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Multi-drug-resistant Staphylococcus aureus infections necessitate novel antibiotic development. D-3263, a transient receptor potential melastatin member 8 (TRPM8) agonist, has potential antineoplastic properties. Here, we reported the antibacterial and antibiofilm activities of D-3263. Minimum inhibitory concentrations (MICs) against S. aureus, Enterococcus faecalis and E. faecium were ≤ 50 µM. D-3263 exhibited bactericidal effects against clinical methicillin-resistant S. aureus (MRSA) and E. faecalis strains at 4× MIC. Subinhibitory D-3263 concentrations effectively inhibited S. aureus and E. faecalis biofilms, with higher concentrations also clearing mature biofilms. Proteomic analysis revealed differential expression of 29 proteins under 1/2 × MIC D-3263, influencing amino acid biosynthesis and carbohydrate metabolism. Additionally, D-3263 enhanced membrane permeability of S. aureus and E. faecalis. Bacterial membrane phospholipids phosphatidylethanolamine (PE), phosphatidylglycerol (PG), and cardiolipin (CL) dose-dependently increased D-3263 MICs. Overall, our data suggested that D-3263 exhibited potent antibacterial and antibiofilm activities against S. aureus by targeting the cell membrane.
Collapse
Affiliation(s)
- Xiaoju Liu
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Yanpeng Xiong
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Renhai Peng
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Yufang Zhang
- Department of Biology, Washington University in St. Louis, 1 Brookings Drive, St Louis, MO, 63130, USA
| | - Shuyu Cai
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Qiwen Deng
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Zhijian Yu
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Zewen Wen
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Zhong Chen
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
| | - Tieying Hou
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
| |
Collapse
|
5
|
Abdulrehman T, Qadri S, Haik Y, Sultan A, Skariah S, Kumar S, Mendoza Z, Yadav KK, Titus A, Khader S. Advances in the targeted theragnostics of osteomyelitis caused by Staphylococcus aureus. Arch Microbiol 2024; 206:288. [PMID: 38834761 DOI: 10.1007/s00203-024-04015-2] [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: 03/31/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
Bone infections caused by Staphylococcus aureus may lead to an inflammatory condition called osteomyelitis, which results in progressive bone loss. Biofilm formation, intracellular survival, and the ability of S. aureus to evade the immune response result in recurrent and persistent infections that present significant challenges in treating osteomyelitis. Moreover, people with diabetes are prone to osteomyelitis due to their compromised immune system, and in life-threatening cases, this may lead to amputation of the affected limbs. In most cases, bone infections are localized; thus, early detection and targeted therapy may prove fruitful in treating S. aureus-related bone infections and preventing the spread of the infection. Specific S. aureus components or overexpressed tissue biomarkers in bone infections could be targeted to deliver active therapeutics, thereby reducing drug dosage and systemic toxicity. Compounds like peptides and antibodies can specifically bind to S. aureus or overexpressed disease markers and combining these with therapeutics or imaging agents can facilitate targeted delivery to the site of infection. The effectiveness of photodynamic therapy and hyperthermia therapy can be increased by the addition of targeting molecules to these therapies enabling site-specific therapy delivery. Strategies like host-directed therapy focus on modulating the host immune mechanisms or signaling pathways utilized by S. aureus for therapeutic efficacy. Targeted therapeutic strategies in conjunction with standard surgical care could be potential treatment strategies for S. aureus-associated osteomyelitis to overcome antibiotic resistance and disease recurrence. This review paper presents information about the targeting strategies and agents for the therapy and diagnostic imaging of S. aureus bone infections.
Collapse
Affiliation(s)
- Tahir Abdulrehman
- eHealth Program, DeGroote School of Business, McMaster University, Hamilton, ON, Canada
- Health Policy, Management and Informatics, Allied Health, Credit Valley Hospital, Mississauga, ON, Canada
| | - Shahnaz Qadri
- School of Pharmacy, Texas A&M University, Kingsville, USA.
| | - Yousef Haik
- Department of Mechanical & Nuclear Engineering, University of Sharjah, Sharjah, UAE.
| | - Ali Sultan
- Department of Immunology & Microbiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Sini Skariah
- Department of Immunology & Microbiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Shourya Kumar
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Zachary Mendoza
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Kamlesh K Yadav
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Anoop Titus
- Department of Preventive Cardiology, Houston Methodist, Houston, TX, USA
| | - Shameer Khader
- School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
6
|
Wang L, Liu L, Zhang C, Yu G, Lin W, Duan X, Xiong Y, Jiang G, Wang J, Liao X. Design, synthesis, anti-infective potency and mechanism study of novel Ru-based complexes containing substituted adamantane as antibacterial agents. Eur J Med Chem 2024; 270:116378. [PMID: 38604098 DOI: 10.1016/j.ejmech.2024.116378] [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: 10/06/2023] [Revised: 03/30/2024] [Accepted: 03/31/2024] [Indexed: 04/13/2024]
Abstract
Infections caused by Staphylococcus aureus (S. aureus) are increasing difficult to treat because this pathogen is easily resistant to antibiotics. However, the development of novel antibacterial agents with high antimicrobial activity and low frequency of resistance remains a huge challenge. Here, building on the coupling strategy, an adamantane moiety was linked to the membrane-active Ru-based structure and then developed three novel metalloantibiotics: [Ru(bpy)2(L)](PF6)2 (Ru1) (bpy = 2,2-bipyridine, L = amantadine modified ligand), [Ru(dmb)2(L)](PF6)2 (Ru2) (dmb = 4,4'-dimethyl-2,2'-bipyridine) and [Ru(dpa)2(L)](PF6)2 (Ru3), (dpa = 2,2'-dipyridylamine). Notably, complex Ru1 was identified to be the best candidate agent, showing greater efficacy against S. aureus than most of clinical antibiotics and low resistance frequencies. Mechanism studies demonstrated that Ru1 could not only increase the permeability of bacterial cell membrane and then caused the leakage of bacterial contents, but also promoted the production of reactive oxygen species (ROS) in bacteria. Importantly, complex Ru1 inhibited the biofilm formation, exotoxin secretion and increased the potency of some clinical used antibiotics. In addition, Ru1 showed low toxic in vivo and excellent anti-infective efficacy in two animal infection model. Thus, Ru-based metalloantibiotic bearing adamantane moiety are promising antibacterial agents, providing a certain research basis for the future antibiotics research.
Collapse
Affiliation(s)
- Liqiang Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China
| | - Lianghong Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, 418000, China
| | - Chunyan Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China
| | - Guangying Yu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China
| | - Wenjing Lin
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China
| | - Xuemin Duan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China
| | - Yanshi Xiong
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China
| | - Guijuan Jiang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China.
| | - Jintao Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China.
| | - Xiangwen Liao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China.
| |
Collapse
|
7
|
Richardson IM, Calo CJ, Ginter EL, Niehaus E, Pacheco KA, Hind LE. Diverse bacteria elicit distinct neutrophil responses in a physiologically relevant model of infection. iScience 2024; 27:108627. [PMID: 38188520 PMCID: PMC10770534 DOI: 10.1016/j.isci.2023.108627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/24/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
An efficient neutrophil response is critical for fighting bacterial infections, which remain a significant global health concern; therefore, modulating neutrophil function could be an effective therapeutic approach. While we have a general understanding of how neutrophils respond to bacteria, how neutrophil function differs in response to diverse bacterial infections remains unclear. Here, we use a microfluidic infection-on-a-chip device to investigate the neutrophil response to four bacterial species: Pseudomonas aeruginosa, Salmonella enterica, Listeria monocytogenes, and Staphylococcus aureus. We find enhanced neutrophil extravasation to L. monocytogenes, a limited overall response to S. aureus, and identify IL-6 as universally important for neutrophil extravasation. Furthermore, we demonstrate a higher percentage of neutrophils generate reactive oxygen species (ROS) when combating gram-negative bacteria versus gram-positive bacteria. For all bacterial species, we found the percentage of neutrophils producing ROS increased following extravasation through an endothelium, underscoring the importance of studying neutrophil function in physiologically relevant models.
Collapse
Affiliation(s)
- Isaac M. Richardson
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| | - Christopher J. Calo
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| | - Eric L. Ginter
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| | - Elise Niehaus
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| | - Kayla A. Pacheco
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| | - Laurel E. Hind
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| |
Collapse
|
8
|
Liu L, Zhao N, Yang K, Liao H, Liu X, Wu Y, Wang Y, Peng X, Wu Y. Proteomic Analysis of Staphylococcus aureus Treated with ShangKeHuangShui. Biol Pharm Bull 2024; 47:292-302. [PMID: 38281773 DOI: 10.1248/bpb.b23-00471] [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: 01/30/2024]
Abstract
Staphylococcus aureus (SAU) stands as the prevailing pathogen in post-traumatic infections, with the emergence of antibiotic resistance presenting formidable treatment hurdles. The pressing need is to explore novel antibiotics to address this challenge. ShangKeHuangShui (SKHS), a patented traditional Chinese herbal formula, has gained widespread use in averting post-traumatic infections, but its biological effects remain incomplete understanding. This study's primary objective was to delve into the antibacterial properties, potential antibacterial compounds within SKHS, and their associated molecular targets. In vitro SKHS antibacterial assays demonstrated that the minimum inhibitory concentration (MIC) was 8.625 mg/mL and the minimum bactericide concentration (MBC) was 17.25 mg/mL. Proteomic analysis based on tandem mass tag (TMT) showed significant changes in the expression level of 246 proteins in SKHS treated group compared to control group, with 79 proteins upregulated and 167 proteins downregulated (>1.5-fold, p < 0.05). Subsequently, thirteen target proteins related to various biological processes and multiple metabolic pathways were selected to conduct parallel reaction monitoring (PRM) and molecular docking screen. In protein tyrosine phosphatase PtpA (ptpA) docking screening, phellodendrine and obacunone can bind to ptpA with the binding energy of - 8.4 and - 8.3 kcal/mol, respectively. This suggests their potential impact on antibacterial activity by modulating the two-component system of SAU. The discovery lays a groundwork for future research endeavors for exploring new antibacterial candidates and elucidating specific active chemical components within SKHS that match target proteins. Further investigations are imperative to unveil the biological effects of these monomers and their potential synergistic actions.
Collapse
Affiliation(s)
- Lichu Liu
- Institute of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine
| | - Na Zhao
- Institute of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine
| | - Kuangyang Yang
- Institute of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine
| | - Honghong Liao
- Institute of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine
| | - Xiaofang Liu
- Institute of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine
| | - Ying Wu
- Laboratory Medicine Center, Foshan Hospital of Traditional Chinese Medicine
| | - Yan Wang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences
| | - Xiao Peng
- Institute of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine
| | - Yuanyan Wu
- Institute of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine
| |
Collapse
|
9
|
Zheng YY, Chung WH, Leung YC, Wong KY. Heterogenous Expression and Purification of Lipid II Flippase from Staphylococcus aureus. Protein Pept Lett 2024; 31:386-394. [PMID: 38967080 PMCID: PMC11348468 DOI: 10.2174/0109298665316374240531113258] [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: 03/28/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Staphylococcus aureus is a common pathogen with strains that are resistant to existing antibiotics. MurJ from S. aureus (SaMurJ), an integral membrane protein functioning as Lipid II flippase, is a potential target for developing new antibacterial agents against this pathogen. Successful expression and purification of this protein shall be useful in the development of drugs against this target. OBJECTIVE In this study, we demonstrated the optimized expression and purification procedures of SaMurJ, identified suitable detergent for extracting and solubilizing the protein, and examined the peptidisc system to generate a detergent-free environment. METHODS SaMurJ fused with N-terminal ten-His tag was expressed without induction. Six detergents were selected for screening the most efficient candidate for extraction and solubilization of the protein. The thermostability of the detergent-solubilized protein was assessed by evaluated temperature incubation. Different ratios of peptidisc bi-helical peptide (NSPr) to SaMurJ were mixed and the on-bead peptidisc assembly method was applied. RESULTS SaMurJ expressed in BL21(DE3) was confirmed by peptide fingerprinting, with a yield of 1 mg SaMurJ per liter culture. DDM was identified as the optimum detergent for solubilization and the nickel affinity column enabled SaMurJ purification with a purity of ~88%. However, NSPr could not stabilize SaMurJ. CONCLUSION The expression and purification of SaMurJ were successful, with high purity and good yield. SaMurJ can be solubilized and stabilized by a DDM-containing buffer.
Collapse
Affiliation(s)
- Yuan Yuan Zheng
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Wai-Hong Chung
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Yun-Chung Leung
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Kwok-Yin Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| |
Collapse
|
10
|
Li X, Hao Y, Yang N, Mao R, Teng D, Wang J. Plectasin: from evolution to truncation, expression, and better druggability. Front Microbiol 2023; 14:1304825. [PMID: 38188573 PMCID: PMC10771296 DOI: 10.3389/fmicb.2023.1304825] [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: 11/01/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Non-computational classical evolution analysis of plectasin and its functional relatives can especially contribute tool value during access to meet requirements for their better druggability in clinical use. Staphylococcus aureus is a zoonotic pathogen that can infect the skin, blood, and other tissues of humans and animals. The impact of pathogens on humans is exacerbated by the crisis of drug resistance caused by the misuse of antibiotics. In this study, we analyzed the evolution of anti-Staphylococcus target functional sequences, designed a series of plectasin derivatives by truncation, and recombinantly expressed them in Pichia pastoris X-33, from which the best recombinant Ple-AB was selected for the druggability study. The amount of total protein reached 2.9 g/L following 120 h of high-density expression in a 5-L fermenter. Ple-AB was found to have good bactericidal activity against gram-positive bacteria, with minimum inhibitory concentration (MIC) values ranging between 2 and 16 μg/mL. It showed good stability and maintained its bactericidal activity during high temperatures, strong acid and alkali environments. Notably, Ple-AB exhibited better druggability, including excellent trypsin resistance, and still possessed approximately 50% of its initial activity following exposure to simulated intestinal fluids for 1 h. In vitro safety testing of Ple-AB revealed low hemolytic activity against mouse erythrocytes and cytotoxicity against murine-derived macrophages. This study successfully realized the high expression of a new antimicrobial peptide (AMP), Ple-AB, in P. pastoris and the establishment of its oral administration as an additive form with high trypsin resistance; the study also revealed its antibacterial properties, indicating that truncation design is a valuable tool for improving druggability and that the candidate Ple-AB may be a novel promising antimicrobial agent.
Collapse
Affiliation(s)
- Xuan Li
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| | - Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Chinese Academy of Agricultural Sciences, Department of Agriculture and Rural Affairs, Beijing, China
| |
Collapse
|
11
|
Wang LQ, Zhang CY, Chen JJ, Lin WJ, Yu GY, Deng LS, Ji XR, Duan XM, Xiong YS, Jiang GJ, Wang JT, Liao XW, Liu LH. Ru-Based Organometallic Agents Bearing Phenyl Hydroxide: Synthesis and Antibacterial Mechanism Study against Staphylococcus aureus. ChemMedChem 2023; 18:e202300306. [PMID: 37527976 DOI: 10.1002/cmdc.202300306] [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: 06/13/2023] [Revised: 07/17/2023] [Accepted: 08/01/2023] [Indexed: 08/03/2023]
Abstract
The development of antimicrobial agents with novel model of actions is a promising strategy to combat multiple resistant bacteria. Here, three ruthenium-based complexes, which acted as potential antimicrobial agents, were synthesized and characterized. Importantly, three complexes all showed strong bactericidal potency against Staphylococcus aureus. In particular, the most active one has a MIC of 6.25 μg/mL. Mechanistic studies indicated that ruthenium complex killed S. aureus by releasing ROS and damaging the integrity of bacterial cell membrane. In addition, the most active complex not only could inhibit the biofilm formation and hemolytic toxin secretion of S. aureus, but also serve as a potential antimicrobial adjuvant as well, which showed synergistic effects with eight traditional antibiotics. Finally, both G. mellonella larva infection model and mouse skin infection model all demonstrated that ruthenium complex also showed significant efficacy against S. aureus in vivo. In summary, our study suggested that ruthenium-based complexes bearing a phenyl hydroxide are promising antimicrobial agents for combating S. aureus.
Collapse
Affiliation(s)
- L Q Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - C Y Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - J J Chen
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - W J Lin
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - G Y Yu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - L S Deng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - X R Ji
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - X M Duan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Y S Xiong
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - G J Jiang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - J T Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - X W Liao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - L H Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, 418000, China
| |
Collapse
|
12
|
Jiang F, Cai C, Wang X, Han S. A dual biomarker-targeting probe enables signal-on surface labeling of Staphylococcus aureus. Bioorg Med Chem Lett 2023; 93:129428. [PMID: 37541632 DOI: 10.1016/j.bmcl.2023.129428] [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: 05/12/2023] [Revised: 07/13/2023] [Accepted: 07/30/2023] [Indexed: 08/06/2023]
Abstract
Imaging or killing of a specific pathogen is of significance for precise therapy. Staphylococcus aureus (S. aureus) is an infectious gram-positive bacteria relying on Sortase A (SrtA) to anchor cell surface protein on peptidoglycan. We herein report signal-on labeling of S. aureus with self-quenched optical probes featuring vancomycin-conjugated SrtA substrate that is flanked by a dabcyl moiety paired with either fluorescein or eosine photosensizer (PS). SrtA-mediated cleavage of the substrate motif releases the dabcyl quencher, leading to covalent labeling of peptidoglycan with fluorescein or PS of restored photophysical property. The dual biomarked-enabled peptidoglycan labeling enables signal-on imaging and effective photodynamic destruction of S. aureus, suggesting a protheranostic approch activatable to SrtA-positive bacteria engaged in myriad diseases.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, the Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361005, China
| | - Chengteng Cai
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, the Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361005, China
| | - Xiumin Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China.
| | - Shoufa Han
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, the Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
13
|
Wang B, Wang H, Lu X, Zheng X, Yang Z. Recent Advances in Electrochemical Biosensors for the Detection of Foodborne Pathogens: Current Perspective and Challenges. Foods 2023; 12:2795. [PMID: 37509887 PMCID: PMC10379338 DOI: 10.3390/foods12142795] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
Foodborne pathogens cause many diseases and significantly impact human health and the economy. Foodborne pathogens mainly include Salmonella spp., Escherichia coli, Staphylococcus aureus, Shigella spp., Campylobacter spp. and Listeria monocytogenes, which are present in agricultural products, dairy products, animal-derived foods and the environment. Various pathogens in many different types of food and water can cause potentially life-threatening diseases and develop resistance to various types of antibiotics. The harm of foodborne pathogens is increasing, necessitating effective and efficient methods for early monitoring and detection. Traditional methods, such as real-time polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA) and culture plate, are time-consuming, labour-intensive and expensive and cannot satisfy the demands of rapid food testing. Therefore, new fast detection methods are urgently needed. Electrochemical biosensors provide consumer-friendly methods to quickly detect foodborne pathogens in food and the environment and achieve extensive accuracy and reproducible results. In this paper, by focusing on various mechanisms of electrochemical transducers, we present a comprehensive overview of electrochemical biosensors for the detection of foodborne pathogens. Furthermore, the review introduces the hazards of foodborne pathogens, risk analysis methods and measures of control. Finally, the review also emphasizes the recent research progress and solutions regarding the use of electrochemical biosensors to detect foodborne pathogens in food and the environment, evaluates limitations and challenges experienced during the development of biosensors to detect foodborne pathogens and discusses future possibilities.
Collapse
Affiliation(s)
- Bo Wang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225009, China
| | - Hang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xubin Lu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Xiangfeng Zheng
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225009, China
| | - Zhenquan Yang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
14
|
Hou Z, Liu L, Wei J, Xu B. Progress in the Prevalence, Classification and Drug Resistance Mechanisms of Methicillin-Resistant Staphylococcus aureus. Infect Drug Resist 2023; 16:3271-3292. [PMID: 37255882 PMCID: PMC10226514 DOI: 10.2147/idr.s412308] [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: 03/13/2023] [Accepted: 05/12/2023] [Indexed: 06/01/2023] Open
Abstract
Staphylococcus aureus is a common human pathogen with a variety of virulence factors, which can cause multiple infectious diseases. In recent decades, due to the constant evolution and the abuse of antibiotics, Staphylococcus aureus was becoming more resistant, the infection rate of MRSA remained high, and clinical treatment of MRSA became more difficult. The genetic diversity of MRSA was mainly represented by the continuous emergence of epidemic strains, resulting in the constant changes of epidemic clones. Different classes of MRSA resulted in different epidemics and resistance characteristics, which could affect the clinical symptoms and treatments. MRSA had also spread from traditional hospitals to community and livestock environments, and the new clones established a relationship between animals and humans, promoting further evolution of MRSA. Since the resistance mechanism of MRSA is very complex, it is important to clarify these resistance mechanisms at the molecular level for the treatment of infectious diseases. We firstly described the diversity of SCCmec elements, and discussed the types of SCCmec, its drug resistance mechanisms and expression regulations. Then, we described how the vanA operon makes Staphylococcus aureus resistant to vancomycin and its expression regulation. Finally, a brief introduction was given to the drug resistance mechanisms of biofilms and efflux pump systems. Analyzing the resistance mechanism of MRSA can help study new anti-infective drugs and alleviate the evolution of MRSA. At the end of the review, we summarized the treatment strategies for MRSA infection, including antibiotics, anti-biofilm agents and efflux pump inhibitors. To sum up, here we reviewed the epidemic characteristics of Staphylococcus aureus, summarized its classifications, drug resistance mechanisms of MRSA (SCCmec element, vanA operon, biofilm and active efflux pump system) and novel therapy strategies, so as to provide a theoretical basis for the treatment of MRSA infection.
Collapse
Affiliation(s)
- Zhuru Hou
- Department of Basic Medicine, Fenyang College of Shanxi Medical University, Fenyang, People’s Republic of China
- Key Laboratory of Lvliang for Clinical Molecular Diagnostics, Fenyang, People’s Republic of China
| | - Ling Liu
- Key Laboratory of Lvliang for Clinical Molecular Diagnostics, Fenyang, People’s Republic of China
- Department of Medical Laboratory Science, Fenyang College of Shanxi Medical University, Fenyang, People’s Republic of China
- Department of Clinical Laboratory, Fenyang Hospital of Shanxi Province, Fenyang, People’s Republic of China
| | - Jianhong Wei
- Department of Basic Medicine, Fenyang College of Shanxi Medical University, Fenyang, People’s Republic of China
| | - Benjin Xu
- Key Laboratory of Lvliang for Clinical Molecular Diagnostics, Fenyang, People’s Republic of China
- Department of Medical Laboratory Science, Fenyang College of Shanxi Medical University, Fenyang, People’s Republic of China
- Department of Clinical Laboratory, Fenyang Hospital of Shanxi Province, Fenyang, People’s Republic of China
| |
Collapse
|
15
|
Nguyen TTT, Nguyen TTT, Nguyen HD, Nguyen TK, Pham PTV, Tran LT, Tran LTT, Tran MH. Integrating in Silico and In Vitro Studies to Screen Anti- Staphylococcus aureus Activity From Vietnamese Ganoderma multiplicatum and Ganoderma sinense. Nat Prod Commun 2023. [DOI: 10.1177/1934578x231167289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Background: Staphylococcus aureus is a nosocomial pathogen responsible for many serious infectious diseases in humans. Finding the anti- S. aureus agents is a time-consuming and costly process. Recently, computational methods have provided a better understanding of the interactions between herbal medicine drug targets to help clinical practitioners rationally design herbal formulae. Methods: In this study, molecular docking simulation was applied to screen a list of natural secondary metabolites from Ganoderma sp. on the protein target S. aureus sortase A. Molecular dynamics models were used to assess the stability of protein–ligand complexes during the first 100 ns. To validate the computational results, 2 Ganoderma species, G. multiplicatum VNKKK1901 and G. sinense VNKKK1902, were tested for antibacterial activity against S. aureus using the disk diffusion method. Results: The results showed that, among the selected compounds, ganosinensin B and ganosinoside A generated the highest binding energy on S. aureus sortase A, and demonstrated strong and stable binding capacity to proteins. In addition, the extracts of G. sinense VNKKK1902 and G. multiplicatum VNKKK1901 were bactericidal, with minimum bactericidal concentration (MBC)/minimum inhibitory concentration (MIC) ratios of 2. Conclusion: Our findings provide the first scientific report on the antibacterial activity of Ganoderma sp., which contain 2 promising compounds, ganosinensin B and ganosinoside A, as potential hits for developing novel drugs capable of supporting treatment of S. aureus infection.
Collapse
Affiliation(s)
- Trang Thi Thu Nguyen
- Faculty of Biology and Biotechnology, University of Science, Ho Chi Minh City Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Trinh Thi Tuyet Nguyen
- Faculty of Biology and Biotechnology, University of Science, Ho Chi Minh City Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Hoang Duc Nguyen
- Faculty of Biology and Biotechnology, University of Science, Ho Chi Minh City Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Tan Khanh Nguyen
- Scientific Management Department, Dong A University, Da Nang city Vietnam
| | - Phu Tran Vinh Pham
- Faculty of Medicine, Dong A University, Hai Chau District, Da Nang City, Vietnam
| | - Linh Thuoc Tran
- Faculty of Biology and Biotechnology, University of Science, Ho Chi Minh City Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Linh Thuy Thi Tran
- Faculty of Pharmacy, Hue University of Medicine and Pharmacy, Hue University, Hue City, Vietnam
| | - Manh Hung Tran
- School of Medicine and Pharmacy, The University of Danang, Da Nang City, Vietnam
| |
Collapse
|
16
|
Wiman E, Zattarin E, Aili D, Bengtsson T, Selegård R, Khalaf H. Development of novel broad-spectrum antimicrobial lipopeptides derived from plantaricin NC8 β. Sci Rep 2023; 13:4104. [PMID: 36914718 PMCID: PMC10011573 DOI: 10.1038/s41598-023-31185-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Bacterial resistance towards antibiotics is a major global health issue. Very few novel antimicrobial agents and therapies have been made available for clinical use during the past decades, despite an increasing need. Antimicrobial peptides have been intensely studied, many of which have shown great promise in vitro. We have previously demonstrated that the bacteriocin Plantaricin NC8 αβ (PLNC8 αβ) from Lactobacillus plantarum effectively inhibits Staphylococcus spp., and shows little to no cytotoxicity towards human keratinocytes. However, due to its limitations in inhibiting gram-negative species, the aim of the present study was to identify novel antimicrobial peptidomimetic compounds with an enhanced spectrum of activity, derived from the β peptide of PLNC8 αβ. We have rationally designed and synthesized a small library of lipopeptides with significantly improved antimicrobial activity towards both gram-positive and gram-negative bacteria, including the ESKAPE pathogens. The lipopeptides consist of 16 amino acids with a terminal fatty acid chain and assemble into micelles that effectively inhibit and kill bacteria by permeabilizing their cell membranes. They demonstrate low hemolytic activity and liposome model systems further confirm selectivity for bacterial lipid membranes. The combination of lipopeptides with different antibiotics enhanced the effects in a synergistic or additive manner. Our data suggest that the novel lipopeptides are promising as future antimicrobial agents, however additional experiments using relevant animal models are necessary to further validate their in vivo efficacy.
Collapse
Affiliation(s)
- Emanuel Wiman
- School of Medical Sciences, Faculty of Medicine and Health, Department of Microbiology, Immunology and Reproductive Science, Örebro University, Örebro, Sweden
| | - Elisa Zattarin
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Daniel Aili
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Torbjörn Bengtsson
- School of Medical Sciences, Faculty of Medicine and Health, Department of Microbiology, Immunology and Reproductive Science, Örebro University, Örebro, Sweden
| | - Robert Selegård
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden.
| | - Hazem Khalaf
- School of Medical Sciences, Faculty of Medicine and Health, Department of Microbiology, Immunology and Reproductive Science, Örebro University, Örebro, Sweden.
| |
Collapse
|
17
|
Seal S, Banerjee N, Mahato R, Kundu T, Sinha D, Chakraborty T, Sinha D, Sau K, Chatterjee S, Sau S. Serine 106 preserves the tertiary structure, function, and stability of a cyclophilin from Staphylococcus aureus. J Biomol Struct Dyn 2023; 41:1479-1494. [PMID: 34967275 DOI: 10.1080/07391102.2021.2021992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
SaCyp, a staphylococcal cyclophilin involved in both protein folding and pathogenesis, has a Ser residue at position 106 and a Trp residue at position 136. While Ser 106 of SaCyp aligned with a cyclosporin A (CsA) binding Ala residue, its Trp 136 aligned with a Trp or a Phe residue of most other cyclophilins. To demonstrate the exact roles of Ser 106 and Trp 136 in SaCyp, we have elaborately studied rCyp[S106A] and rCyp[W136A], two-point mutants of a recombinant SaCyp (rCyp) harboring an Ala substitution at positions 106 and 136, respectively. Of the mutants, rCyp[W136A] showed the rCyp-like CsA binding affinity and peptidyl-prolyl cis-trans isomerase (PPIase) activity. Conversely, the PPIase activity, CsA binding affinity, stability, tertiary structure, surface hydrophobicity, and Trp accessibility of rCyp[S106A] notably differed from those of rCyp. The computational experiments also reveal that the structure, dimension, and fluctuation of SaCyp are not identical to those of SaCyp[S106A]. Furthermore, Ser at position 106 of SaCyp, compared to Ala at the same position, formed a higher number of non-covalent bonds with CsA. Collectively, Ser 106 is an indispensable residue for SaCyp that keeps its tertiary structure, function, and stability intact.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Soham Seal
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
| | - Nilanjan Banerjee
- Department of Biophysics, Bose Institute, Kolkata, West Bengal, India
| | - Rohit Mahato
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
| | - Tanmoy Kundu
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
| | - Debabrata Sinha
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
| | | | - Debasmita Sinha
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
| | - Keya Sau
- Department of Biotechnology, Haldia Institute of Technology, Haldia, West Bengal, India
| | | | - Subrata Sau
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
| |
Collapse
|
18
|
Lima A, Carolina Barbosa Caetano A, Hurtado Castillo R, Gonçalves Dos Santos R, Lucas Neres Rodrigues D, de Jesus Sousa T, Kato RB, Vinicius Canário Viana M, Cybelle Pinto Gomide A, Figueira Aburjaile F, Tiwari S, Jaiswal A, Gala-García A, Seyffert N, Luiz de Paula Castro T, Brenig B, Matiuzzi da Costa M, Maria Seles Dorneles E, Le Loir Y, Azevedo V. Comparative genomic analysis of ovine and other host associated isolates of Staphylococcus aureus exhibit the important role of mobile genetic elements and virulence factors in host adaptation. Gene 2023; 855:147131. [PMID: 36539044 DOI: 10.1016/j.gene.2022.147131] [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: 09/15/2022] [Revised: 12/01/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Staphylococcus aureus is the main etiological agent of mastitis in small ruminants worldwide. This disease has a difficult cure and possible relapse, leading to significant economic losses in production, milk quality and livestock. This study performed comparative genomic analyses between 73 S. aureus genomes from different hosts (human, bovine, pig and others). This work isolated and sequenced 12 of these genomes from ovine. This study contributes to the knowledge of genomic specialization and the role of specific genes in establishing infection in ovine mastitis-associated S. aureus. The genomes of S. aureus isolated from sheep maintained a higher representation when grouped with clonal complexes 130 and 133. The genomes showed high genetic similarity, the species pan-genome consisting of 4200 genes (central = 2008, accessory = 1559 and unique = 634). Among these, 277 unique genes were related to the genomes isolated from sheep, with 39.6 % as hypothetical proteins, 6.4 % as phages, 6.4 % as toxins, 2.9 % as transporters, and 44.7 % as related to other proteins. Furthermore, at the pathogen level, they showed 80 genes associated with virulence factors and 19 with antibiotic resistance shared in almost all isolates. Although S. aureus isolated from ovine showed susceptibility to antimicrobials in vitro, ten genes were predicted to be associated with antibiotic inactivation and efflux pump, suggesting resistance to gentamicin and penicillin. This work may contribute to identifying genes acquired by horizontal transfer and their role in host adaptation, virulence, bacterial resistance, and characterization of strains affecting ovine.
Collapse
Affiliation(s)
- Alessandra Lima
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Raquel Hurtado Castillo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Diego Lucas Neres Rodrigues
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Thiago de Jesus Sousa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo Bentes Kato
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Anne Cybelle Pinto Gomide
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flavia Figueira Aburjaile
- Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sandeep Tiwari
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Programa de Pós-graduação em Imunologia, Instituto De Biologia, Universidade Federal da Bahia, Salvador, BA, Brasil.; Programa de Pós-graduação em Microbiologia, Instituto De Biologia, Universidade Federal da Bahia, Salvador, BA, Brasil
| | - Arun Jaiswal
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alfonso Gala-García
- Faculdade de Odontologia, Universidade Federal da Bahia, Salvador, BA, Brazil
| | - Núbia Seyffert
- Programa de Pós-graduação em Imunologia, Instituto De Biologia, Universidade Federal da Bahia, Salvador, BA, Brasil
| | - Thiago Luiz de Paula Castro
- Programa de Pós-graduação em Imunologia, Instituto De Biologia, Universidade Federal da Bahia, Salvador, BA, Brasil
| | - Bertram Brenig
- Institute of Veterinary Medicine, University of Göttingen, Burckhardtweg 2, Göttingen, Germany
| | - Mateus Matiuzzi da Costa
- Laboratório de Microbiologia e Imunologia Animal, Campus Ciências Agrárias, Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, Pernambuco, Brazil
| | - Elaine Maria Seles Dorneles
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Medicina Veterinária, Universidade Federal de Lavras, Lavras, Minas Gerais, Brazil
| | - Yves Le Loir
- Institut National de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Paris, France
| | - Vasco Azevedo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
19
|
Kumar G, Kiran Tudu A. Tackling multidrug-resistant Staphylococcus aureus by natural products and their analogues acting as NorA efflux pump inhibitors. Bioorg Med Chem 2023; 80:117187. [PMID: 36731248 DOI: 10.1016/j.bmc.2023.117187] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
Staphylococcus aureus (S. aureus) is a pathogen responsible for various community and hospital-acquired infections with life-threatening complications like bacteraemia, endocarditis, meningitis, liver abscess, and spinal cord epidural abscess. Antibiotics have been used to treat microbial infections since the introduction of penicillin in 1940. In recent decades, the abuse and misuse of antibiotics in humans, animals, plants, and fungi, including the treatment of non-microbial diseases, have led to the rapid emergence of multidrug-resistant pathogens with increased virulence. Bacteria have developed several complementary mechanisms to avoid the effects of antibiotics. These mechanisms include chemical transformations and enzymatic inactivation of antibiotics, modification of antibiotics' target site, and reduction of intracellular antibiotics concentration by changes in membrane permeability or by the overexpression of efflux pumps (EPs). The strategy to check antibiotic resistance includes synthesis of the antibiotic analogues, or antibiotics are given in combination with the adjuvant. The inhibitors of multidrug EPs are considered promising alternative therapeutic options with the potential to revive the effects of antibiotics and reduce bacterial virulence. Natural products played a vital role in drug discovery and significantly contributed to the area of infectious diseases. Also, natural products provide lead compounds that sometimes need modification based on structural and biological properties to meet the drug criteria. This review discusses natural products and their derived compounds as NorA efflux pump inhibitors (EPIs).
Collapse
Affiliation(s)
- Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, Telangana 500037, India.
| | - Asha Kiran Tudu
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, Telangana 500037, India
| |
Collapse
|
20
|
Zhang X, Xiong T, Gao L, Wang Y, Liu L, Tian T, Shi Y, Zhang J, Zhao Z, Lu D, Luo P, Zhang W, Cheng P, Jing H, Gou Q, Zeng H, Yan D, Zou Q. Extracellular fibrinogen-binding protein released by intracellular Staphylococcus aureus suppresses host immunity by targeting TRAF3. Nat Commun 2022; 13:5493. [PMID: 36123338 PMCID: PMC9484707 DOI: 10.1038/s41467-022-33205-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 09/08/2022] [Indexed: 11/09/2022] Open
Abstract
Many pathogens secrete effectors to hijack intracellular signaling regulators in host immune cells to promote pathogenesis. However, the pathogenesis of Staphylococcus aureus secretory effectors within host cells is unclear. Here, we report that Staphylococcus aureus secretes extracellular fibrinogen-binding protein (Efb) into the cytoplasm of macrophages to suppress host immunity. Mechanistically, RING finger protein 114, a host E3 ligase, mediates K27-linked ubiquitination of Efb at lysine 71, which facilitates the recruitment of tumor necrosis factor receptor associated factor (TRAF) 3. The binding of Efb to TRAF3 disrupts the formation of the TRAF3/TRAF2/cIAP1 (cellular-inhibitor-of-apoptosis-1) complex, which mediates K48-ubiquitination of TRAF3 to promote degradation, resulting in suppression of the inflammatory signaling cascade. Additionally, the Efb K71R mutant loses the ability to inhibit inflammation and exhibits decreased pathogenicity. Therefore, our findings identify an unrecognized mechanism of Staphylococcus aureus to suppress host defense, which may be a promising target for developing effective anti-Staphylococcus aureus immunomodulators. Staphylococcus aureus secrete numerous effectors to evade or inhibit the host immune response, yet the mechanism underlying the effectors ability to manipulate the signalling pathways of macrophages remain unclear. Authors utilise in vitro and in vivo models to explore the role of extracellular fibrinogen-binding protein (Efb) in immune response modulation and pathogenicity.
Collapse
Affiliation(s)
- Xiaokai Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Tingrong Xiong
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Lin Gao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Yu Wang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China.,Department of Basic Courses, NCO School, Third Military Medical University, Shijiazhuang, 050081, China
| | - Luxuan Liu
- College of Medicine, Southwest Jiaotong University, Chengdu, 610083, China
| | - Tian Tian
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Yun Shi
- Institute of Biopharmaceutical Research, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Haiming Jing
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Qiang Gou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China.
| | - Dapeng Yan
- Department of Immunology, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity & Shanghai Public Health Clinical Center, Fudan University, Shanghai, 200032, China.
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
21
|
Morita Y, Saito M, Rangel-Moreno J, Franchini AM, Owen JR, Martinez JC, Daiss JL, de Mesy Bentley KL, Kates SL, Schwarz EM, Muthukrishnan G. Systemic IL-27 administration prevents abscess formation and osteolysis via local neutrophil recruitment and activation. Bone Res 2022; 10:56. [PMID: 36028492 PMCID: PMC9418173 DOI: 10.1038/s41413-022-00228-7] [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: 02/20/2022] [Revised: 05/20/2022] [Accepted: 06/15/2022] [Indexed: 01/07/2023] Open
Abstract
Interleukin-27 is a pleiotropic cytokine whose functions during bacterial infections remain controversial, and its role in patients with S. aureus osteomyelitis is unknown. To address this knowledge gap, we completed a clinical study and observed elevated serum IL-27 levels (20-fold higher, P < 0.05) in patients compared with healthy controls. Remarkably, IL-27 serum levels were 60-fold higher in patients immediately following septic death than in uninfected patients (P < 0.05), suggesting a pathogenic role of IL-27. To test this hypothesis, we evaluated S. aureus osteomyelitis in WT and IL-27Rα-/- mice with and without exogenous IL-27 induction by intramuscular injection of rAAV-IL-27p28 or rAAV-GFP, respectively. We found that IL-27 was induced at the surgical site within 1 day of S. aureus infection of bone and was expressed by M0, M1 and M2 macrophages and osteoblasts but not by osteoclasts. Unexpectedly, exogenous IL-27p28 (~2 ng·mL-1 in serum) delivery ameliorated soft tissue abscesses and peri-implant bone loss during infection, accompanied by enhanced local IL-27 expression, significant accumulation of RORγt+ neutrophils at the infection site, a decrease in RANK+ cells, and compromised osteoclast formation. These effects were not observed in IL-27Rα-/- mice compared with WT mice, suggesting that IL-27 is dispensable for immunity but mediates redundant immune and bone cell functions during infection. In vitro studies and bulk RNA-seq of infected tibiae showed that IL-27 increased nos1, nos2, il17a, il17f, and rorc expression but did not directly stimulate chemotaxis. Collectively, these results identify a novel phenomenon of IL-27 expression by osteoblasts immediately following S. aureus infection of bone and suggest a protective role of systemic IL-27 in osteomyelitis.
Collapse
Affiliation(s)
- Yugo Morita
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Motoo Saito
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Anthony M Franchini
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - John R Owen
- Department of Orthopedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - John C Martinez
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - John L Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Stephen L Kates
- Department of Orthopedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
22
|
Sherchand SP, Adhikari RP, Muthukrishnan G, Kanipakala T, Owen JR, Xie C, Aman MJ, Proctor RA, Schwarz EM, Kates SL. Evidence of Neutralizing and Non-Neutralizing Anti-Glucosaminidase Antibodies in Patients With S. Aureus Osteomyelitis and Their Association With Clinical Outcome Following Surgery in a Clinical Pilot. Front Cell Infect Microbiol 2022; 12:876898. [PMID: 35923804 PMCID: PMC9339635 DOI: 10.3389/fcimb.2022.876898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/20/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus osteomyelitis remains a very challenging condition; recent clinical studies have shown infection control rates following surgery/antibiotics to be ~60%. Additionally, prior efforts to produce an effective S. aureus vaccine have failed, in part due to lack of knowledge of protective immunity. Previously, we demonstrated that anti-glucosaminidase (Gmd) antibodies are protective in animal models but found that only 6.7% of culture-confirmed S. aureus osteomyelitis patients in the AO Clinical Priority Program (AO-CPP) Registry had basal serum levels (>10 ng/ml) of anti-Gmd at the time of surgery (baseline). We identified a small subset of patients with high levels of anti-Gmd antibodies and adverse outcomes following surgery, not explained by Ig class switching to non-functional isotypes. Here, we aimed to test the hypothesis that clinical cure following surgery is associated with anti-Gmd neutralizing antibodies in serum. Therefore, we first optimized an in vitro assay that quantifies recombinant Gmd lysis of the M. luteus cell wall and used it to demonstrate the 50% neutralizing concentration (NC50) of a humanized anti-Gmd mAb (TPH-101) to be ~15.6 μg/ml. We also demonstrated that human serum deficient in anti-Gmd antibodies can be complemented by TPH-101 to achieve the same dose-dependent Gmd neutralizing activity as purified TPH-101. Finally, we assessed the anti-Gmd physical titer and neutralizing activity in sera from 11 patients in the AO-CPP Registry, who were characterized into four groups post-hoc. Group 1 patients (n=3) had high anti-Gmd physical and neutralizing titers at baseline that decreased with clinical cure of the infection over time. Group 2 patients (n=3) had undetectable anti-Gmd antibodies throughout the study and adverse outcomes. Group 3 (n=3) had high titers +/- neutralizing anti-Gmd at baseline with adverse outcomes. Group 4 (n=2) had low titers of non-neutralizing anti-Gmd at baseline with delayed high titers and adverse outcomes. Collectively, these findings demonstrate that both neutralizing and non-neutralizing anti-Gmd antibodies exist in S. aureus osteomyelitis patients and that screening for these antibodies could have a value for identifying patients in need of passive immunization prior to surgery. Future prospective studies to test the prognostic value of anti-Gmd antibodies to assess the potential of passive immunization with TPH-101 are warranted.
Collapse
Affiliation(s)
| | | | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | | | - John R. Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - M. Javad Aman
- Integrated BioTherapeutics, Inc., Rockville, MD, United States
| | - Richard A. Proctor
- Departments of Medical Microbiology/Immunology and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
23
|
Weldick PJ, Wang A, Halbus AF, Paunov VN. Emerging nanotechnologies for targeting antimicrobial resistance. NANOSCALE 2022; 14:4018-4041. [PMID: 35234774 DOI: 10.1039/d1nr08157h] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Antimicrobial resistance is a leading cause of mortality worldwide. Without newly approved antibiotics and antifungals being brought to the market, resistance is being developed to the ones currently available to clinicians. The reason is the applied evolutionary pressure to bacterial and fungal species due to the wide overuse of common antibiotics and antifungals in clinical practice and agriculture. Biofilms harbour antimicrobial-resistant subpopulations, which make their antimicrobial treatment even more challenging. Nanoparticle-based technologies have recently been shown to successfully overcome antimicrobial resistance in both planktonic and biofilms phenotypes. This results from the combination of novel nanomaterial research and classic antimicrobial therapies which promise to deliver a whole new generation of high-performance active nanocarrier systems. This review discusses the latest developments of promising nanotechnologies with applications against resistant pathogens and evaluates their potential and feasibility for use in novel antimicrobial therapies.
Collapse
Affiliation(s)
- Paul J Weldick
- Department of Chemistry and Biochemistry, University of Hull, Hull, HU6 7RX, UK
| | - Anheng Wang
- Department of Chemistry and Biochemistry, University of Hull, Hull, HU6 7RX, UK
| | - Ahmed F Halbus
- Department of Chemistry, College of Science, University of Babylon, Hilla, Iraq
| | - Vesselin N Paunov
- Department of Chemistry, Nazarbayev University, Kabanbay Baryr Ave. 53, Nur-sultan city, 010000, Kazakhstan.
| |
Collapse
|
24
|
Xiang H, Yang P, Wang L, Li J, Wang T, Xue J, Wang D, Ma H. Isovitexin Is a Direct Inhibitor of Staphylococcus aureus Coagulase. J Microbiol Biotechnol 2021; 31:1350-1357. [PMID: 34409949 PMCID: PMC9706020 DOI: 10.4014/jmb.2105.05013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/28/2021] [Accepted: 08/17/2021] [Indexed: 12/15/2022]
Abstract
Staphylococcus aureus (S. aureus) is a major pathogen that causes human pneumonia, leading to significant morbidity and mortality. S. aureus coagulase (Coa) triggers the polymerization of fibrin by activating host prothrombin, which then converts fibrinogen to fibrin and contributes to S. aureus pathogenesis and persistent infection. In our research, we demonstrate that isovitexin, an active traditional Chinese medicine component, can inhibit the coagulase activity of Coa but does not interfere with the growth of S. aureus. Furthermore, we show through thermal shift and fluorescence quenching assays that isovitexin directly binds to Coa. Dynamic simulation and structure-activity relationship analyses suggest that V191 and P268 are key amino acid residues responsible for the binding of isovitexin to Coa. Taken together, these data indicate that isovitexin is a direct Coa inhibitor and a promising candidate for drug development against S. aureus infection.
Collapse
Affiliation(s)
- Hua Xiang
- College of Animal Medicine, Jilin Agricultural University, Changchun 130118, P.R. China,College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, P.R. China,The Key Laboratory of New Veterinary Drug Research and Development of Jilin Province, Jilin Agricultural University, Changchun 130118, P.R. China
| | - Panpan Yang
- College of Basic Medical Science, Jilin University, Changchun 130012, P.R. China
| | - Li Wang
- College of Animal Science, Jilin University, Changchun 130062, P.R. China
| | - Jiaxin Li
- College of Animal Science, Jilin University, Changchun 130062, P.R. China
| | - Tiedong Wang
- College of Animal Science, Jilin University, Changchun 130062, P.R. China
| | - Junze Xue
- College of Animal Medicine, Jilin Agricultural University, Changchun 130118, P.R. China,College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, P.R. China,The Key Laboratory of New Veterinary Drug Research and Development of Jilin Province, Jilin Agricultural University, Changchun 130118, P.R. China
| | - Dacheng Wang
- College of Animal Science, Jilin University, Changchun 130062, P.R. China,Corresponding author Phone: +86-431-84532812 E-mail:
| | - Hongxia Ma
- College of Animal Medicine, Jilin Agricultural University, Changchun 130118, P.R. China,College of Life Science, Jilin Agricultural University, Changchun 130118, P.R. China,The Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, P.R. China,Corresponding author Phone: +86-431-84532812 E-mail:
| |
Collapse
|
25
|
Molecular insights into mechanisms of GPCR hijacking by Staphylococcus aureus. Proc Natl Acad Sci U S A 2021; 118:2108856118. [PMID: 34663701 DOI: 10.1073/pnas.2108856118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 11/18/2022] Open
Abstract
Atypical chemokine receptor 1 (ACKR1) is a G protein-coupled receptor (GPCR) targeted by Staphylococcus aureus bicomponent pore-forming leukotoxins to promote bacterial growth and immune evasion. Here, we have developed an integrative molecular pharmacology and structural biology approach in order to characterize the effect of leukotoxins HlgA and HlgB on ACKR1 structure and function. Interestingly, using cell-based assays and native mass spectrometry, we found that both components HlgA and HlgB compete with endogenous chemokines through a direct binding with the extracellular domain of ACKR1. Unexpectedly, hydrogen/deuterium exchange mass spectrometry analysis revealed that toxin binding allosterically modulates the intracellular G protein-binding domain of the receptor, resulting in dissociation and/or changes in the architecture of ACKR1-Gαi1 protein complexes observed in living cells. Altogether, our study brings important molecular insights into the initial steps of leukotoxins targeting a host GPCR.
Collapse
|
26
|
Novel ocotillol-derived lactone derivatives: design, synthesis, bioactive evaluation, SARs and preliminary antibacterial mechanism. Mol Divers 2021; 26:2103-2120. [PMID: 34661800 DOI: 10.1007/s11030-021-10318-z] [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: 05/11/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
A new series of ocotillol-derived lactone derivatives were designed and synthesized to consider their antibacterial activity, structure-activity relationships (SARs), antibacterial mechanism and in vivo antibacterial efficacy. Compound 6d, which exhibited broad antibacterial spectrum, was found to be the most active with minimum inhibitory concentrations (MICs) of 1-2 μg/mL against Gram-positive bacteria and 8-16 μg/mL against Gram-negative bacteria. The subsequent synergistic antibacterial tests displayed that 6d had the ability to improve the susceptibility of MRSA USA300, B. subtilis 168, and E. coli DH5α to kanamycin and chloramphenicol. This active molecule 6d also induced bacterial resistance more slowly than norfloxacin and kanamycin. Furthermore, compound 6d was membrane active and low toxic against mammalian cells, and it could rapidly inhibit the growth of MRSA and E. coli and did not obviously trigger bacterial resistance. Compound 6d also displayed strong in vivo antibacterial activity against S. aureus RN4220 in murine corneal infection models. Additionally, absorption, distribution, metabolism, and excretion properties of this type of compounds have shown drug-likeness with good oral absorption and moderate blood-brain barrier permeability. The obtained results demonstrated that ocotillol-derived compounds are a promising class of antibacterial agents worthy of further study.
Collapse
|
27
|
Muthukrishnan G, Beck CA, Owen JR, Xie C, Kates SL, Daiss JL. Serum antibodies against Staphylococcus aureus can prognose treatment success in patients with bone infections. J Orthop Res 2021; 39:2169-2176. [PMID: 33325051 PMCID: PMC8286088 DOI: 10.1002/jor.24955] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/18/2020] [Accepted: 12/14/2020] [Indexed: 02/04/2023]
Abstract
Prognosing life-threatening orthopedic infections caused by Staphylococcus aureus remains a major clinical challenge. To address this, we developed a multiplex assay to assess the humoral immune proteome against S. aureus in patients with musculoskeletal infections. We found initial evidence that antibodies against some antigens (autolysins: Amd, Gmd; secreted immunotoxins: CHIPS, SCIN, Hla) were associated with protection, whereas antibodies against the iron-regulated surface determinant (Isd) proteins (IsdA, IsdB, IsdH) were aligned with adverse outcomes. To formally test this, we analyzed antibody levels and 1-year clinical outcomes of 194 patients with confirmed S. aureus bone infections (AO Trauma Clinical Priority Program [CPP] Bone Infection Registry). A staggering 20.6% of the enrolled patients experienced adverse clinical outcomes (arthrodesis, reinfection, amputation, and septic death) after 1-year. At enrollment, anti-S. aureus immunoglobulin G (IgG) levels in patients with adverse outcomes were 1.35-fold lower than those in patients whose infections were successfully controlled (p < 0.0001). Overall, there was a 51%-69% reduction in adverse outcome risk for every 10-fold increase in initial IgG concentration against Gmd, Amd, IsdH, CHIPS, SCIN, and Hla (p < 0.05). Notably, anti-IsdB antibodies remained elevated in patients with adverse outcomes; for every 10-fold change in the ratio of circulating anti-Isd to anti-Atl IgG at enrollment, there was a trending 2.6-fold increased risk (odds ratio = 2.555) of an adverse event (p = 0.105). Moreover, antibody increases over time correlated with adverse outcomes and decreases with positive outcomes. These studies demonstrate the potential of the humoral immune response against S. aureus as a prognostic indicator for assessing treatment success and identifying patients requiring additional interventions.
Collapse
Affiliation(s)
- Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Christopher A. Beck
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA,Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - John R. Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - John L. Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA,Corresponding Author: John L. Daiss, Ph.D., Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, 601 Elmwood Avenue, Box 651, Rochester, NY, 14642,
| |
Collapse
|
28
|
Milewska S, Niemirowicz-Laskowska K, Siemiaszko G, Nowicki P, Wilczewska AZ, Car H. Current Trends and Challenges in Pharmacoeconomic Aspects of Nanocarriers as Drug Delivery Systems for Cancer Treatment. Int J Nanomedicine 2021; 16:6593-6644. [PMID: 34611400 PMCID: PMC8487283 DOI: 10.2147/ijn.s323831] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Nanotherapy is a part of nanomedicine that involves nanoparticles as carriers to deliver drugs to target locations. This novel targeting approach has been found to resolve various problems, especially those associated with cancer treatment. In nanotherapy, the carrier plays a crucial role in handling many of the existing challenges, including drug protection before early-stage degradations of active substances, allowing them to reach targeted cells and overcome cell resistance mechanisms. The present review comprises the following sections: the first part presents the introduction of pharmacoeconomics as a branch of healthcare economics, the second part covers various beneficial aspects of the use of nanocarriers for in vitro, in vivo, and pre- and clinical studies, as well as discussion on drug resistance problem and present solutions to overcome it. In the third part, progress in drug manufacturing and optimization of the process of nanoparticle synthesis were discussed. Finally, pharmacokinetic and toxicological properties of nanoformulations due to up-to-date studies were summarized. In this review, the most recent developments in the field of nanotechnology's economic impact, particularly beneficial applications in medicine were presented. Primarily focus on cancer treatment, but also discussion on other fields of application, which are strongly associated with cancer epidemiology and treatment, was made. In addition, the current limitations of nanomedicine and its huge potential to improve and develop the health care system were presented.
Collapse
Affiliation(s)
- Sylwia Milewska
- Department of Experimental Pharmacology, Medical University of Bialystok, Bialystok, 15-361, Poland
| | | | | | - Piotr Nowicki
- Department of Experimental Pharmacology, Medical University of Bialystok, Bialystok, 15-361, Poland
| | | | - Halina Car
- Department of Experimental Pharmacology, Medical University of Bialystok, Bialystok, 15-361, Poland
| |
Collapse
|
29
|
Wang YJ, Wang W, You ZY, Liu XX. Observation of synergistic antibacterial properties of prodigiosin from Serratia marcescens jx-1 with metal ions in clinical isolates of Staphylococcus aureus. Prep Biochem Biotechnol 2021; 52:344-350. [PMID: 34289781 DOI: 10.1080/10826068.2021.1944201] [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] [Indexed: 10/20/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections are a major global health problem, and novel and effective antimicrobial drugs are urgently required to combat this life-threatening pathogen. Prodigiosin (PG) is a bacterial secondary metabolite with excellent anticancer and antibacterial properties. However, little is known about the antibacterial function of PG against MRSA. Therefore, the antibacterial efficacy of PG alone and PG in combination with different metal ions against clinic isolates of MRSA and methicillin-sensitive S. aureus (MSSA) strain was evaluated in the present study. The minimum inhibitory concentration of PG against both MRSA and MSSA was 0.25 μg/mL. However, 0.1 μg/mL PG showed a stronger inhibitory effect on MSSA cell growth (47.12%) than on MRSA cell growth (35.87%). Surprisingly, we observed a significant difference (p < 0.01) in membrane integrity between PG-treated MRSA and MSSA using the propidium iodide staining assay. Further, we found that in combination with PG, Zn2+, Al3+, and Cu2+ showed synergistic antibacterial effects against MRSA and MSSA. Our results could increase the current knowledge regarding the efficacy of PG in inhibiting the growth of different types of S. aureus clinical isolates and also offer a novel strategy for developing efficient antibacterial agents.
Collapse
Affiliation(s)
- Yu-Jie Wang
- College of Biological, Chemical Sciences and Engineering, Jiaxing University, Jiaxing, China
| | - Wei Wang
- Clinical Laboratory of First Hospital of Jiaxing, Jiaxing, China
| | - Zhong-Yu You
- College of Biological, Chemical Sciences and Engineering, Jiaxing University, Jiaxing, China
| | - Xiao-Xia Liu
- College of Biological, Chemical Sciences and Engineering, Jiaxing University, Jiaxing, China
| |
Collapse
|
30
|
Giurazza R, Mazza MC, Andini R, Sansone P, Pace MC, Durante-Mangoni E. Emerging Treatment Options for Multi-Drug-Resistant Bacterial Infections. Life (Basel) 2021; 11:life11060519. [PMID: 34204961 PMCID: PMC8229628 DOI: 10.3390/life11060519] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
Antimicrobial resistance (AMR) remains one of the top public health issues of global concern. Among the most important strategies for AMR control there is the correct and appropriate use of antibiotics, including those available for the treatment of AMR pathogens. In this article, after briefly reviewing the most important and clinically relevant multi-drug-resistant bacteria and their main resistance mechanisms, we describe the emerging antimicrobial options for both MDR Gram-positive cocci and Gram-negative bacilli, including recently marketed agents, molecules just approved or under evaluation and rediscovered older antibiotics that have regained importance due to their antimicrobial spectrum. Specifically, emerging options for Gram-positive cocci we reviewed include ceftaroline, ceftobiprole, tedizolid, dalbavancin, and fosfomycin. Emerging treatment options for Gram-negative bacilli we considered comprise ceftolozane-tazobactam, ceftazidime-avibactam, meropenem-vaborbactam, imipenem-relebactam, aztreonam-avibactam, minocycline, fosfomycin, eravacycline, plazomicin, and cefiderocol. An exciting scenario is opening today with the long awaited growing availability of novel molecules for the treatment of AMR bacteria. Knowledge of mechanisms of action and resistance patterns allows physicians to increasingly drive antimicrobial treatment towards a precision medicine approach. Strict adherence to antimicrobial stewardship practices will allow us to preserve the emerging antimicrobials for our future.
Collapse
Affiliation(s)
- Roberto Giurazza
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, Internal Medicine Section, Piazzale Ettore Ruggieri snc, 80131 Naples, Italy; (R.G.); (M.C.M.)
- Department of Woman, Child and General & Specialized Surgery, University of Campania ‘L. Vanvitelli’, Piazza Miraglia, 80138 Naples, Italy; (P.S.); (M.C.P.)
| | - Maria Civita Mazza
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, Internal Medicine Section, Piazzale Ettore Ruggieri snc, 80131 Naples, Italy; (R.G.); (M.C.M.)
- Department of Woman, Child and General & Specialized Surgery, University of Campania ‘L. Vanvitelli’, Piazza Miraglia, 80138 Naples, Italy; (P.S.); (M.C.P.)
| | - Roberto Andini
- Unit of Infectious and Transplant Medicine, AORN Ospedali dei Colli-Monaldi Hospital, Piazzale Ettore Ruggieri snc, 80131 Naples, Italy;
| | - Pasquale Sansone
- Department of Woman, Child and General & Specialized Surgery, University of Campania ‘L. Vanvitelli’, Piazza Miraglia, 80138 Naples, Italy; (P.S.); (M.C.P.)
| | - Maria Caterina Pace
- Department of Woman, Child and General & Specialized Surgery, University of Campania ‘L. Vanvitelli’, Piazza Miraglia, 80138 Naples, Italy; (P.S.); (M.C.P.)
| | - Emanuele Durante-Mangoni
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, Internal Medicine Section, Piazzale Ettore Ruggieri snc, 80131 Naples, Italy; (R.G.); (M.C.M.)
- Unit of Infectious and Transplant Medicine, AORN Ospedali dei Colli-Monaldi Hospital, Piazzale Ettore Ruggieri snc, 80131 Naples, Italy;
- Correspondence: ; Tel.: +39-081-7062475; Fax: +39-081-7702645
| |
Collapse
|
31
|
Morita Y, Masters EA, Schwarz EM, Muthukrishnan G. Interleukin-27 and Its Diverse Effects on Bacterial Infections. Front Immunol 2021; 12:678515. [PMID: 34079555 PMCID: PMC8165262 DOI: 10.3389/fimmu.2021.678515] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/27/2021] [Indexed: 02/03/2023] Open
Abstract
Innate and adaptive immune responses against pathogens are known to be carefully orchestrated by specific cytokines that initiate and down regulate immune cell functions from the initial infection through tissue repair and homeostasis. However, some cytokines, including interleukin-27, are expressed at multiple phases of the infection, such that their pro and anti-inflammatory functions have been difficult to interpret. As elucidation of specific cytokine functions throughout infection is central to our understanding of protective vs. susceptible immunity and return to homeostasis vs. prolonged inflammation leading to septic shock, here we review the literature on IL-27 signaling and the various functions of this heterodimeric ligand member of the IL-12 cytokine family. Canonically, IL-27 is produced by antigen-presenting cells, and is thought of as an immunostimulatory cytokine due to its capacity to induce Th1 differentiation. However, many studies have also identified various immunosuppressive effects of IL-27 signaling, including suppression of Th17 differentiation and induction of co-inhibitory receptors on T cells. Thus, the exact role of IL-27 in the context of infectious diseases remains a topic of debate and active research. Additionally, as recent interest has focused on clinical management of acute vs. chronic infections, and life-threatening "cytokine storm" from sepsis, we propose a hypothetical model to explain the biphasic role of IL-27 during the early and late phases of immune responses to reconcile its known pro and anti-inflammatory functions, which could be therapeutically regulated to improve patient outcomes of infection.
Collapse
Affiliation(s)
- Yugo Morita
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | - Elysia A. Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
32
|
Fisher JF, Mobashery S. β-Lactams against the Fortress of the Gram-Positive Staphylococcus aureus Bacterium. Chem Rev 2021; 121:3412-3463. [PMID: 33373523 PMCID: PMC8653850 DOI: 10.1021/acs.chemrev.0c01010] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The biological diversity of the unicellular bacteria-whether assessed by shape, food, metabolism, or ecological niche-surely rivals (if not exceeds) that of the multicellular eukaryotes. The relationship between bacteria whose ecological niche is the eukaryote, and the eukaryote, is often symbiosis or stasis. Some bacteria, however, seek advantage in this relationship. One of the most successful-to the disadvantage of the eukaryote-is the small (less than 1 μm diameter) and nearly spherical Staphylococcus aureus bacterium. For decades, successful clinical control of its infection has been accomplished using β-lactam antibiotics such as the penicillins and the cephalosporins. Over these same decades S. aureus has perfected resistance mechanisms against these antibiotics, which are then countered by new generations of β-lactam structure. This review addresses the current breadth of biochemical and microbiological efforts to preserve the future of the β-lactam antibiotics through a better understanding of how S. aureus protects the enzyme targets of the β-lactams, the penicillin-binding proteins. The penicillin-binding proteins are essential enzyme catalysts for the biosynthesis of the cell wall, and understanding how this cell wall is integrated into the protective cell envelope of the bacterium may identify new antibacterials and new adjuvants that preserve the efficacy of the β-lactams.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| |
Collapse
|
33
|
Muthukrishnan G, Wallimann A, Rangel-Moreno J, Bentley KLDM, Hildebrand M, Mys K, Kenney HM, Sumrall ET, Daiss JL, Zeiter S, Richards RG, Schwarz EM, Moriarty TF. Humanized Mice Exhibit Exacerbated Abscess Formation and Osteolysis During the Establishment of Implant-Associated Staphylococcus aureus Osteomyelitis. Front Immunol 2021; 12:651515. [PMID: 33815412 PMCID: PMC8012494 DOI: 10.3389/fimmu.2021.651515] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/02/2021] [Indexed: 12/17/2022] Open
Abstract
Staphylococcus aureus is the predominant pathogen causing osteomyelitis. Unfortunately, no immunotherapy exists to treat these very challenging and costly infections despite decades of research, and numerous vaccine failures in clinical trials. This lack of success can partially be attributed to an overreliance on murine models where the immune correlates of protection often diverge from that of humans. Moreover, S. aureus secretes numerous immunotoxins with unique tropism to human leukocytes, which compromises the targeting of immune cells in murine models. To study the response of human immune cells during chronic S. aureus bone infections, we engrafted non-obese diabetic (NOD)-scid IL2Rγnull (NSG) mice with human hematopoietic stem cells (huNSG) and analyzed protection in an established model of implant-associated osteomyelitis. The results showed that huNSG mice have increases in weight loss, osteolysis, bacterial dissemination to internal organs, and numbers of Staphylococcal abscess communities (SACs), during the establishment of implant-associated MRSA osteomyelitis compared to NSG controls (p < 0.05). Flow cytometry and immunohistochemistry demonstrated greater human T cell numbers in infected versus uninfected huNSG mice (p < 0.05), and that T-bet+ human T cells clustered around the SACs, suggesting S. aureus-mediated activation and proliferation of human T cells in the infected bone. Collectively, these proof-of-concept studies underscore the utility of huNSG mice for studying an aggressive form of S. aureus osteomyelitis, which is more akin to that seen in humans. We have also established an experimental system to investigate the contribution of specific human T cells in controlling S. aureus infection and dissemination.
Collapse
Affiliation(s)
- Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | - Alexandra Wallimann
- AO Research Institute Davos, Davos, Switzerland.,Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | | | - Karen Mys
- AO Research Institute Davos, Davos, Switzerland
| | - H Mark Kenney
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | | | - John L Daiss
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | | | | | - Edward M Schwarz
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States.,Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | | |
Collapse
|
34
|
A narrative review of single-nucleotide polymorphism detection methods and their application in studies of Staphylococcus aureus. JOURNAL OF BIO-X RESEARCH 2021. [DOI: 10.1097/jbr.0000000000000071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
35
|
Tsegaye MM, Chouhan G, Fentie M, Tyagi P, Nand P. Therapeutic Potential of Green Synthesized Metallic Nanoparticles against Staphylococcus aureus. Curr Drug Res Rev 2021; 13:172-183. [PMID: 33634763 DOI: 10.2174/2589977513666210226123920] [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: 05/16/2020] [Revised: 09/08/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The recent treatment challenges posed by the widespread emergence of pathogenic Multidrug-Resistant (MDR) bacterial strains are a cause of huge health troubles worldwide. Infections caused by MDR organisms are associated with longer period of hospitalization, increased mortality, and inflated healthcare costs. Staphylococcus aureus is one of these MDR organisms identified as an urgent threat to human health by the World Health Organization. Infections caused by S. aureus may range from simple cutaneous infestations to life threatening bacteremia. S. aureus infections get easily escalated in severely ill, hospitalized and or immunocompromised patients with incapacitated immune system. Also, in HIV-positive patients S. aureus ranks amongst one of the most common comorbidities where it can further worsen a patient's health condition. At present anti-staphylococcal therapy is reliant typically on chemotherapeutics that are gathering resistance and pose unfavorable side-effects. Thus, newer drugs are required that can bridge these shortcomings and aid effective control against S. aureus. OBJECTIVE In this review, we summarize drug resistance exhibited by S. aureus and lacunae in current anti-staphylococcal therapy, nanoparticles as an alternative therapeutic modality. The focus lays on various green synthesized nanoparticles, their mode of action and application as potent antibacterial compounds against S. aureus. CONCLUSION Use of nanoparticles as anti-bacterial drugs has gained momentum in recent past and green synthesized nanoparticles, which involves microorganisms and plants or their byproducts for synthesis of nanoparticles offer a potent, as well as environment friendly solution in warfare against MDR bacte.
Collapse
Affiliation(s)
- Meron Moges Tsegaye
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh. India
| | - Garima Chouhan
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh. India
| | - Molla Fentie
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh. India
| | - Priya Tyagi
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh. India
| | - Parma Nand
- School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh. India
| |
Collapse
|
36
|
Pakki E, Tayeb R, Usmar U, Ridwan IA, Muslimin L. Effect of orally administered combination of Caulerpa racemosa and Eleutherine americana (Aubl) Merr extracts on phagocytic activity of macrophage. Res Pharm Sci 2020; 15:401-409. [PMID: 33312218 PMCID: PMC7714016 DOI: 10.4103/1735-5362.293518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/25/2020] [Accepted: 07/11/2020] [Indexed: 11/13/2022] Open
Abstract
Background and purpose: Polysaccharide sulfate is a major active phytochemical constituent of Caulerpa racemosa, whereas the Eleutherine americana (Aubl) Merr has antioxidant properties. The aim of this research was to investigate the combined effect of polysaccharide sulfate that was isolated from C. racemosa and E. americana on the macrophage activity. Experimental approach: The phenolic contents and antioxidant activities of E. americana extracts in water and various ethanol concentrations were studied using the Folin-Ciocalteu and 2,2-diphenyl-1-picryl-hydrazyl- hydrate (DPPH) methods, respectively. Polysaccharide sulfate was isolated from C. racemosa by precipitation method. To assess the macrophage activity, mice were treated orally for 14 days with either a combination of polysaccharide sulfate and E. americana 96% ethanol extract at a specific ratio or with each extract alone. Macrophages were isolated and the phagocytic activity was measured by assessing the ability of the macrophages to phagocytose latex particles and nitric oxide (NO) levels were assessed using a colorimetric assay. Findings / Results: The E. americana crude extract in water exhibited the highest yield (13.04%), compared with the extract in 96% ethanol, which had the highest phenolic content (6.37 ± 0.16 mg/g gallic acid equivalent) and the strongest antioxidant activity (IC50, 22.63 ± 1.09 μg/mL). The combination of extracts, when both extracts were administered at 65:65 mg/kg BW, resulted in the highest increases in phagocytosis activity (62.73 ± 5.77%) and NO levels (16.43 ± 1.37 μmol/L). Conclusion and implications: The results of this study confirmed the non-specific immunostimulant properties of the combination of polysaccharide sulfate and E. americana and justified their use in traditional medicine. The observed increase in macrophage activity appeared to be correlated with the increased ability of mice to fight infection.
Collapse
Affiliation(s)
- Ermina Pakki
- Faculty of Pharmacy, Hasanuddin University, Perintis Kemerdekaan KM 10, Makassar, Indonesia, 90245
| | - Rosany Tayeb
- Faculty of Pharmacy, Hasanuddin University, Perintis Kemerdekaan KM 10, Makassar, Indonesia, 90245
| | - Usmar Usmar
- Faculty of Pharmacy, Hasanuddin University, Perintis Kemerdekaan KM 10, Makassar, Indonesia, 90245
| | - Ismul Azham Ridwan
- Faculty of Pharmacy, Hasanuddin University, Perintis Kemerdekaan KM 10, Makassar, Indonesia, 90245
| | - Lukman Muslimin
- Department of Pharmaceutical Chemistry, Sekolah Tinggi Ilmu Farmasi Makassar, Makassar, Indonesia 90241
| |
Collapse
|
37
|
Gao Y, Chen Y, Cao Y, Mo A, Peng Q. Potentials of nanotechnology in treatment of methicillin-resistant Staphylococcus aureus. Eur J Med Chem 2020; 213:113056. [PMID: 33280899 DOI: 10.1016/j.ejmech.2020.113056] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 02/05/2023]
Abstract
Abuse of antibiotics has led to the emergence of drug-resistant pathogens. Methicillin-resistant Staphylococcus aureus (MRSA) was reported just two years after the clinical use of methicillin, which can cause severe infections with high morbidity and mortality in both community and hospital. The treatment of MRSA infection is greatly challenging since it has developed the resistance to almost all types of antibiotics. As such, it is of great significance and importance to develop novel therapeutic approaches. The fast development of nanotechnology provides a promising solution to this dilemma. Functional nanomaterials and nanoparticles can act either as drug carriers or as antibacterial agents for antibacterial therapy. Herein, we aim to provide a comprehensive understanding of the drug resistance mechanisms of MRSA and discuss the potential applications of some functionalized nanomaterials in anti-MRSA therapy. Also, the concerns and possible solutions for the nanomaterials-based anti-MRSA therapy are discussed.
Collapse
Affiliation(s)
- Yujie Gao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuan Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yubin Cao
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Anchun Mo
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
38
|
Kates SL, Owen JR, Beck CA, Xie C, Muthukrishnan G, Daiss JL, Schwarz EM. Lack of Humoral Immunity Against Glucosaminidase Is Associated with Postoperative Complications in Staphylococcus aureus Osteomyelitis. J Bone Joint Surg Am 2020; 102:1842-1848. [PMID: 32858560 PMCID: PMC9018051 DOI: 10.2106/jbjs.20.00029] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Glucosaminidase (Gmd) is known to be a protective antigen in animal models of Staphylococcus aureus osteomyelitis. We compared the endogenous anti-Gmd antibody levels in sera of patients with culture-confirmed S. aureus bone infections to their sera at 1 year after operative treatment of the infection. METHODS A novel global biospecimen registry of 297 patients with deep-wound culture-confirmed S. aureus osteomyelitis was analyzed to assess relationships between baseline anti-Gmd serum titers (via custom Luminex assay), known host risk factors for infection, and 1-year postoperative clinical outcomes (e.g., infection control, inconclusive, refracture, persistent infection, septic nonunion, amputation, and septic death). RESULTS All patients had measurable humoral immunity against some S. aureus antigens, but only 20 patients (6.7%; p < 0.0001) had high levels of anti-Gmd antibodies (>10 ng/mL) in serum at baseline. A subset of 194 patients (65.3%) who completed 1 year of follow-up was divided into groups based on anti-Gmd level: low (<1 ng/mL, 54 patients; 27.8%), intermediate (<10 ng/mL, 122 patients; 62.9%), and high (>10 ng/mL, 18 patients; 9.3%), and infection control rates were 40.7%, 50.0%, and 66.7%, respectively. The incidence of adverse outcomes in these groups was 33.3%, 16.4%, and 11.1%, respectively. Assessing anti-Gmd level as a continuous variable showed a 60% reduction in adverse-event odds (p = 0.04) for every tenfold increase in concentration. No differences in patient demographics, body mass index of >40 kg/m, diabetes status, age of ≥70 years, male sex, Charlson Comorbidity Index of >1, or Cierny-Mader host type were observed between groups, and these risk factors were not associated with adverse events. Patients with low anti-Gmd titer demonstrated a significant 2.68-fold increased odds of adverse outcomes (p = 0.008). CONCLUSIONS Deficiency in circulating anti-Gmd antibodies was associated serious adverse outcomes following operative treatment of S. aureus osteomyelitis. At 1 year, high levels of anti-Gmd antibodies were associated with a nearly 3-fold increase in infection-control odds. Additional prospective studies clarifying Gmd immunization for osteomyelitis are needed. LEVEL OF EVIDENCE Prognostic Level IV. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - John R. Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Christopher A. Beck
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | | | - John L. Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
39
|
Shang L, Li J, Song C, Nina Z, Li Q, Chou S, Wang Z, Shan A. Hybrid Antimicrobial Peptide Targeting Staphylococcus aureus and Displaying Anti-infective Activity in a Murine Model. Front Microbiol 2020; 11:1767. [PMID: 33042031 PMCID: PMC7516806 DOI: 10.3389/fmicb.2020.01767] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Broad-spectrum antimicrobial peptides (AMPs) kill bacteria indiscriminately, increasing the possibility of an ecological imbalance in the microbiota. To solve this problem, new types of AMPs, which kill pathogenic bacteria without breaking the micro-ecological balance of the body, were proposed. Here, we successfully designed a targeting AMP, S2, which is a fusion peptide composed of a species-specific targeting domain and broad-spectrum AMP domain. In the current study, S2 showed specific killing activity against Staphylococcus aureus, and almost no resistance induced compared to penicillin. Mechanism studies indicated that S2 killed S. aureus by destroying the bacterial membrane. Meanwhile, S2 possessed excellent salt-tolerance properties and biocompatibility. Importantly, S2 exhibited perfect treatment efficacy against an S. aureus subcutaneous infection model and remained nontoxic. In conclusion, this study provides a promising strategy for designing specific AMPs against growing bacterial infections.
Collapse
Affiliation(s)
- Lu Shang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Jiawei Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Chunsheng Song
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Zaytseva Nina
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Qiuke Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Shuli Chou
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Zhihua Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
40
|
Muthukrishnan G, Soin S, Beck CA, Grier A, Brodell JD, Lee CC, Ackert-Bicknell CL, Lee FEH, Schwarz EM, Daiss JL. A Bioinformatic Approach to Utilize a Patient's Antibody-Secreting Cells against Staphylococcus aureus to Detect Challenging Musculoskeletal Infections. Immunohorizons 2020; 4:339-351. [PMID: 32571786 DOI: 10.4049/immunohorizons.2000024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/04/2020] [Indexed: 01/22/2023] Open
Abstract
Noninvasive diagnostics for Staphylococcus aureus musculoskeletal infections (MSKI) remain challenging. Abs from newly activated, pathogen-specific plasmablasts in human blood, which emerge during an ongoing infection, can be used for diagnosing and tracking treatment response in diabetic foot infections. Using multianalyte immunoassays on medium enriched for newly synthesized Abs (MENSA) from Ab-secreting cells, we assessed anti-S. aureus IgG responses in 101 MSKI patients (63 culture-confirmed S. aureus, 38 S. aureus-negative) and 52 healthy controls. MENSA IgG levels were assessed for their ability to identify the presence and type of S. aureus MSKI using machine learning and multivariate receiver operating characteristic curves. Eleven S. aureus-infected patients were presented with prosthetic joint infections, 15 with fracture-related infections, 5 with native joint septic arthritis, 15 with diabetic foot infections, and 17 with suspected orthopedic infections in the soft tissue. Anti-S. aureus MENSA IgG levels in patients with non-S. aureus infections and healthy controls were 4-fold (***p = 0.0002) and 8-fold (****p < 0.0001) lower, respectively, compared with those with culture-confirmed S. aureus infections. Comparison of MENSA IgG responses among S. aureus culture-positive patients revealed Ags predictive of active MSKI (IsdB, SCIN, Gmd) and Ags predictive of MSKI type (IsdB, IsdH, Amd, Hla). When combined, IsdB, IsdH, Gmd, Amd, SCIN, and Hla were highly discriminatory of S. aureus MSKI (area under the ROC curve = 0.89 [95% confidence interval 0.82-0.93, p < 0.01]). Collectively, these results demonstrate the feasibility of a bioinformatic approach to use a patient's active immune proteome against S. aureus to diagnose challenging MSKI.
Collapse
Affiliation(s)
- Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Sandeep Soin
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Christopher A Beck
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642.,Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642
| | - Alex Grier
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| | - James D Brodell
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Charles C Lee
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Cheryl L Ackert-Bicknell
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopedics, University of Colorado Denver, Denver, CO 80045; and
| | - Frances Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642.,Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| | - John L Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642; .,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
41
|
Sampaio de Oliveira KB, Leite ML, Rodrigues GR, Duque HM, da Costa RA, Cunha VA, de Loiola Costa LS, da Cunha NB, Franco OL, Dias SC. Strategies for recombinant production of antimicrobial peptides with pharmacological potential. Expert Rev Clin Pharmacol 2020; 13:367-390. [PMID: 32357080 DOI: 10.1080/17512433.2020.1764347] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION The need to develop new drugs for the control of pathogenic microorganisms has redoubled efforts to prospect for antimicrobial peptides (AMPs) from natural sources and to characterize its structure and function. These molecules present a broad spectrum of action against different microorganisms and frequently present promiscuous action, with anticancer and immunomodulatory activities. Furthermore, AMPs can be used as biopharmaceuticals in the treatment of hospital-acquired infections and other serious diseases with relevant social and economic impacts.Areas covered: The low yield and the therefore difficult extraction and purification process in AMPs are problems that limit their industrial application and scientific research. Thus, optimized heterologous expression systems were developed to significantly boost AMP yields, allow high efficiency in purification and structural optimization for the increase of therapeutic activity.Expert opinion: This review provides an update on recent developments in the recombinant production of ribosomal and non-ribosomal synthesis of AMPs and on strategies to increase the expression of genes encoding AMPs at the transcriptional and translational levels and regulation of the post-translational modifications. Moreover, there are detailed reports of AMPs that have already reached marketable status or are in the pipeline under advanced stages of preclinical testing.
Collapse
Affiliation(s)
- Kamila Botelho Sampaio de Oliveira
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília , Brasília, Brazil
| | - Michel Lopes Leite
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília , Brasília, Brazil
| | - Gisele Regina Rodrigues
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília , Brasília, Brazil
| | - Harry Morales Duque
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília , Brasília, Brazil
| | - Rosiane Andrade da Costa
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília , Brasília, Brazil
| | - Victor Albuquerque Cunha
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília , Brasília, Brazil
| | - Lorena Sousa de Loiola Costa
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília , Brasília, Brazil
| | - Nicolau Brito da Cunha
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília , Brasília, Brazil
| | - Octavio Luiz Franco
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília , Brasília, Brazil.,Universidade de Brasília, Pós-graduação em Patologia Molecular, Campus Darcy Ribeiro , Brasília, Brazil.,S-Inova Biotech, Pós-graduação em Biotecnologia, Universidade Católica Dom Bosco , Campo Grande, Mato Grosso do Sul, Brazil
| | - Simoni Campos Dias
- Centro de Análises Proteômicas e Bioquímicas, Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília , Brasília, Brazil.,Universidade de Brasília, Pós-graduação em Biologia Animal, Campus Darcy Ribeiro , Brasília, Brazil
| |
Collapse
|
42
|
Chen G, Bai Y, Li Z, Wang F, Fan X, Zhou X. Bacterial extracellular vesicle-coated multi-antigenic nanovaccines protect against drug-resistant Staphylococcus aureus infection by modulating antigen processing and presentation pathways. Am J Cancer Res 2020; 10:7131-7149. [PMID: 32641983 PMCID: PMC7330855 DOI: 10.7150/thno.44564] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/20/2020] [Indexed: 01/31/2023] Open
Abstract
Background: Vaccination provides an alternative to antibiotics in addressing drug-resistant Staphylococcus aureus (S. aureus) infection. However, vaccine potency is often limited by a lack of antigenic breadth and a demand on the generation of antibody responses alone. Methods: In this study, bacterial extracellular vesicles (EVs) coating indocyanine green (ICG)-loaded magnetic mesoporous silica nanoparticles (MSN) were constructed as multi-antigenic vaccines (EV/ICG/MSN) with the ability to modulate antigen presentation pathways in dendritic cells (DCs) to induce cellular immune responses. Results: Exposing the EV/ICG/MSNs to a laser could promote DC maturation and enhance the proteasome-dependent antigen presentation pathway by facilitating endolysosomal escape, improving proteasome activity, and elevating MHC-I expression. Immunization by EV/ICG/MSNs with laser irradiation in vivo triggered improved CD8+ T cell responses while maintaining CD4+ T cell responses and humoral immunity. In addition, in vivo tracking data revealed that the vaccine could be efficiently transported from the injection site into lymph nodes. Skin infection experiments showed that the vaccine not only prevented and treated superficial infection but also decreased bacterial invasiveness, thus strongly suggesting that EV/ICG/MSNs were effective in preventing complications resulting from the introduction of S. aureus infections. Conclusion: This multi-antigenic nanovaccine-based modulation of antigen presentation pathways provides an effective strategy against drug-resistant S. aureus infection.
Collapse
|
43
|
Scolari IR, Páez PL, Musri MM, Petiti JP, Torres A, Granero GE. Rifampicin loaded in alginate/chitosan nanoparticles as a promising pulmonary carrier against Staphylococcus aureus. Drug Deliv Transl Res 2020; 10:1403-1417. [PMID: 32363536 DOI: 10.1007/s13346-019-00705-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
This study aims to explore the antimicrobial activity of rifampicin (RIF) and ascorbic acid (ASC) co-loaded into alginate (ALG)/chitosan (CS) nanoparticles (RIF/ASC NPs) and tested for their antibacterial activity against several strains of methicillin-sensitive Staphylococcus aureus (MSSA) and methicillin-resistant Staphylococcus aureus (MRSA). Also, the present research focused on exploring the possible antibacterial mechanism of action of these RIF/ASC NPs, which demonstrated a significant biocide activity against the S. aureus strains with minimum inhibitory concentrations (MIC) between 2- and 8-fold lower than those one exhibited with the free antibiotic RIF. The proposed antimicrobial mechanism of action of the RIF/ASC NPs seems to be the result of collaborative effects between NPs and the RIF/ASC antibiotic combination. Moreover, results indicated that the functionalized RIF/ASC NP surface played a crucial role on the processes of NP adhesion into the bacterial surface, the alterations on the cell membrane integrity, and the cell uptake of the RIF/ASC antibiotic into bacteria. Further, the in vivo lung deposition pattern of empty NPs labeled (NPs-FITC) with isothiocyanate fluorescein in rats was investigated post intratracheal instillation of NPs. In summary, findings from this work show that our novel designed engineered RIF/ASC co-loaded NPs could be a suitable system for antibiotic lung administration with promising perspectives for effective treatments of pulmonary intracellular infections for those known antibiotics that are losing effectiveness due to antimicrobial resistance problems. Graphical Abstract.
Collapse
Affiliation(s)
- I R Scolari
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA - CONICET) and Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - P L Páez
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA - CONICET) and Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - M M Musri
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Córdoba, Argentina.,Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - J P Petiti
- INICSA, CONICET Centro de Microscopía Electrónica, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - A Torres
- INICSA, CONICET Centro de Microscopía Electrónica, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - G E Granero
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA - CONICET) and Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina.
| |
Collapse
|
44
|
Monteiro AFM, Scotti MT, Speck-Planche A, Barros RPC, Scotti L. In Silico Studies for Bacterystic Evaluation against Staphylococcus aureus of 2-Naphthoic Acid Analogues. Curr Top Med Chem 2020; 20:293-304. [DOI: 10.2174/1568026619666191206111742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 09/01/2019] [Accepted: 09/10/2019] [Indexed: 01/27/2023]
Abstract
Background:
Staphylococcus aureus is a gram-positive spherical bacterium commonly present in
nasal fossae and in the skin of healthy people; however, in high quantities, it can lead to complications that
compromise health. The pathologies involved include simple infections, such as folliculitis, acne, and delay in
the process of wound healing, as well as serious infections in the CNS, meninges, lung, heart, and other areas.
Aim:
This research aims to propose a series of molecules derived from 2-naphthoic acid as a bioactive in the
fight against S. aureus bacteria through in silico studies using molecular modeling tools.
Methods:
A virtual screening of analogues was done in consideration of the results that showed activity according
to the prediction model performed in the KNIME Analytics Platform 3.6, violations of the Lipinski
rule, absorption rate, cytotoxicity risks, energy of binder-receptor interaction through molecular docking, and
the stability of the best profile ligands in the active site of the proteins used (PDB ID 4DXD and 4WVG).
Results:
Seven of the 48 analogues analyzed showed promising results for bactericidal action against S.
aureus.
Conclusion:
It is possible to conclude that ten of the 48 compounds derived from 2-naphthoic acid presented
activity based on the prediction model generated, of which seven presented no toxicity and up to one violation
to the Lipinski rule.
Collapse
Affiliation(s)
| | - Marcus Tullius Scotti
- Federal University of Paraíba, Health Science Center, 50670-910, Joao Pessoa, PB, Brazil
| | - Alejandro Speck-Planche
- Department of Chemistry, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Trubetskaya Str., 8, b. 2, 119992, Moscow, Russian Federation
| | | | - Luciana Scotti
- Federal University of Paraíba, Health Science Center, 50670-910, Joao Pessoa, PB, Brazil
| |
Collapse
|
45
|
Fursova K, Sorokin A, Sokolov S, Dzhelyadin T, Shulcheva I, Shchannikova M, Nikanova D, Artem'eva O, Zinovieva N, Brovko F. Virulence Factors and Phylogeny of Staphylococcus aureus Associated With Bovine Mastitis in Russia Based on Genome Sequences. Front Vet Sci 2020; 7:135. [PMID: 32270001 PMCID: PMC7111254 DOI: 10.3389/fvets.2020.00135] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/24/2020] [Indexed: 12/28/2022] Open
Abstract
Staphylococcus aureus is a causative agent of different infectious processes, food poisoning, and autoimmune disorders. The horizontal transfer of pathogenic strains can occur from animal to human under both house and farm conditions, and the spread of strains with antibiotic resistance is an existing problem. In addition to the spread of antibiotic-resistant strains in clinics, this problem also exists in veterinary medicine. It is especially important to monitor antibiotic resistance on farms where antibiotics are the standard treatment of animals, which may trigger the spread of antibiotic-resistant strains among animals and to the human population, and these strains can also be distributed in milk products produced by these farms (milk, cheese, and butter). In this work, we investigated 21 S. aureus isolates using whole-genome sequence analysis and tried to establish a relationship between these isolates with the development of bovine mastitis in seven regions of Western Russia. An S. aureus virulence profile was identified. We identified two groups of S. aureus associated with subclinical mastitis, namely, the enterotoxin-positive and enterotoxin-negative groups. The most prevalent factor associated with bovine mastitis in Russia was cytotoxins, including hemolysins and leukocidins. Multidrug resistance strains were investigated, and antibiotic resistance genes were identified. We identified S. aureus ST 97 type as the most common type in the regions in Western Russia. To the best of our knowledge, this is the first in-depth study of a range S. aureus isolates originating from cattle infections in Russia.
Collapse
Affiliation(s)
- Ksenia Fursova
- Laboratory of Immunochemistry, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Pushchino, Russia
| | - Anatoly Sorokin
- Laboratory of Cell Genome Functioning Mechanisms, Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino, Russia
| | - Sergey Sokolov
- Laboratory of Plasmid Biology, G.K. Skryabin Institute of Biochemistry & Physiology of Microorganisms of the Russian Academy of Sciences, Pushchino, Russia
| | - Timur Dzhelyadin
- Laboratory of Cell Genome Functioning Mechanisms, Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino, Russia
| | - Irina Shulcheva
- Laboratory of Immunochemistry, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Pushchino, Russia
| | - Margarita Shchannikova
- Laboratory of Immunochemistry, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Pushchino, Russia
| | - Daria Nikanova
- Laboratory of Microbiology, L.K. Ernst Federal Science Center for Animal Husbandry, Moscow, Russia
| | - Olga Artem'eva
- Laboratory of Microbiology, L.K. Ernst Federal Science Center for Animal Husbandry, Moscow, Russia
| | - Natalia Zinovieva
- Laboratory of Microbiology, L.K. Ernst Federal Science Center for Animal Husbandry, Moscow, Russia
| | - Fedor Brovko
- Laboratory of Immunochemistry, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Pushchino, Russia.,Laboratory of Microbiology, L.K. Ernst Federal Science Center for Animal Husbandry, Moscow, Russia
| |
Collapse
|
46
|
Fisher JF, Mobashery S. Constructing and deconstructing the bacterial cell wall. Protein Sci 2020; 29:629-646. [PMID: 31747090 PMCID: PMC7021008 DOI: 10.1002/pro.3737] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022]
Abstract
The history of modern medicine cannot be written apart from the history of the antibiotics. Antibiotics are cytotoxic secondary metabolites that are isolated from Nature. The antibacterial antibiotics disproportionately target bacterial protein structure that is distinct from eukaryotic protein structure, notably within the ribosome and within the pathways for bacterial cell-wall biosynthesis (for which there is not a eukaryotic counterpart). This review focuses on a pre-eminent class of antibiotics-the β-lactams, exemplified by the penicillins and cephalosporins-from the perspective of the evolving mechanisms for bacterial resistance. The mechanism of action of the β-lactams is bacterial cell-wall destruction. In the monoderm (single membrane, Gram-positive staining) pathogen Staphylococcus aureus the dominant resistance mechanism is expression of a β-lactam-unreactive transpeptidase enzyme that functions in cell-wall construction. In the diderm (dual membrane, Gram-negative staining) pathogen Pseudomonas aeruginosa a dominant resistance mechanism (among several) is expression of a hydrolytic enzyme that destroys the critical β-lactam ring of the antibiotic. The key sensing mechanism used by P. aeruginosa is monitoring the molecular difference between cell-wall construction and cell-wall deconstruction. In both bacteria, the resistance pathways are manifested only when the bacteria detect the presence of β-lactams. This review summarizes how the β-lactams are sensed and how the resistance mechanisms are manifested, with the expectation that preventing these processes will be critical to future chemotherapeutic control of multidrug resistant bacteria.
Collapse
Affiliation(s)
- Jed F. Fisher
- Department of Chemistry and BiochemistryUniversity of Notre DameSouth BendIndiana
| | - Shahriar Mobashery
- Department of Chemistry and BiochemistryUniversity of Notre DameSouth BendIndiana
| |
Collapse
|
47
|
Bengtsson T, Selegård R, Musa A, Hultenby K, Utterström J, Sivlér P, Skog M, Nayeri F, Hellmark B, Söderquist B, Aili D, Khalaf H. Plantaricin NC8 αβ exerts potent antimicrobial activity against Staphylococcus spp. and enhances the effects of antibiotics. Sci Rep 2020; 10:3580. [PMID: 32107445 PMCID: PMC7046733 DOI: 10.1038/s41598-020-60570-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/13/2020] [Indexed: 02/07/2023] Open
Abstract
The use of conventional antibiotics has substantial clinical efficacy, however these vital antimicrobial agents are becoming less effective due to the dramatic increase in antibiotic-resistant bacteria. Novel approaches to combat bacterial infections are urgently needed and bacteriocins represent a promising alternative. In this study, the activities of the two-peptide bacteriocin PLNC8 αβ were investigated against different Staphylococcus spp. The peptide sequences of PLNC8 α and β were modified, either through truncation or replacement of all L-amino acids with D-amino acids. Both L- and D-PLNC8 αβ caused rapid disruption of lipid membrane integrity and were effective against both susceptible and antibiotic resistant strains. The D-enantiomer was stable against proteolytic degradation by trypsin compared to the L-enantiomer. Of the truncated peptides, β1–22, β7–34 and β1–20 retained an inhibitory activity. The peptides diffused rapidly (2 min) through the bacterial cell wall and permeabilized the cell membrane, causing swelling with a disorganized peptidoglycan layer. Interestingly, sub-MIC concentrations of PLNC8 αβ substantially enhanced the effects of different antibiotics in an additive or synergistic manner. This study shows that PLNC8 αβ is active against Staphylococcus spp. and may be developed as adjuvant in combination therapy to potentiate the effects of antibiotics and reduce their overall use.
Collapse
Affiliation(s)
- Torbjörn Bengtsson
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, SE-70362, Sweden
| | - Robert Selegård
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, SE-70362, Sweden.,Division of Molecular Physics, Department of Physics, Chemistry and Biology (IFM), Linköping University, Linköping, SE-58183, Sweden
| | - Amani Musa
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, SE-70362, Sweden
| | - Kjell Hultenby
- Department of Laboratory Medicine, Division of Clinical Research Centre, Karolinska Institutet, Stockholm, SE-14186, Sweden
| | - Johanna Utterström
- Division of Molecular Physics, Department of Physics, Chemistry and Biology (IFM), Linköping University, Linköping, SE-58183, Sweden
| | | | | | - Fariba Nayeri
- PEAS Research Institute, Department of Infection Control, Linköping, SE-58273, Sweden
| | - Bengt Hellmark
- Department of Clinical Microbiology, Örebro University Hospital, Örebro, SE-70185, Sweden
| | - Bo Söderquist
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, SE-70362, Sweden.,Department of Clinical Microbiology, Örebro University Hospital, Örebro, SE-70185, Sweden
| | - Daniel Aili
- Division of Molecular Physics, Department of Physics, Chemistry and Biology (IFM), Linköping University, Linköping, SE-58183, Sweden
| | - Hazem Khalaf
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, SE-70362, Sweden.
| |
Collapse
|
48
|
Lee JK, Mereuta L, Luchian T, Park Y. Antimicrobial peptide HPA3NT3-A2 effectively inhibits biofilm formation in mice infected with drug-resistant bacteria. Biomater Sci 2019; 7:5068-5083. [PMID: 31555777 DOI: 10.1039/c9bm01051c] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bacterial biofilms formed through secretion of extracellular polymeric substances (EPS) have been implicated in many serious infections and can increase antibiotic resistance by a factor of more than 1000. Here, we examined the abilities of the antimicrobial peptide HPA3NT3-A2 to inhibit and reduce biofilm formation, eliminate EPS, and suppress inflammation in mice infected with clinical isolates of drug-resistant Pseudomonas aeruginosa strains. HPA3NT3-A2 was developed from a desirable analogue peptide, HPA3NT3, derived from residues 2-20 of the Helicobacter pylori ribosomal protein L1. HPA3NT3-A2 showed stronger activity against planktonic cells (MIC: 8 μM) compared to ciprofloxacin or tobramycin (>512 μM), and a favorable minimum biofilm inhibition and elimination concentration. This peptide also neutralized LPS; decreased levels of EPS; inhibited the production of pro-inflammatory cytokines in the lung, kidney, and spleen; decreased white blood cell counts; and increased survival among infected mice.
Collapse
Affiliation(s)
- Jong-Kook Lee
- Research Center for Proteinaceous Materials (RCPM), Chosun University, Gwangju 501-759, Korea
| | | | | | | |
Collapse
|
49
|
Hilgeroth A, Yasrebi K, Suzen S, Hertlein T, Ohlsen K, Lalk M. Antibacterial Evaluation of Novel Substituted Cycloheptaindoles in Staphylococcus and Enterococcus Strains. Med Chem 2019; 15:833-839. [DOI: 10.2174/1573406415666190208170126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/16/2019] [Accepted: 01/19/2019] [Indexed: 11/22/2022]
Abstract
Background:
Due to emerging resistances against antibiotics there is a strong need to
find novel antibacterial agents with a novel structure to prevent early resistance developments.
Objective:
Bisindole compounds with antibacterial activities which formally result from the reaction
of an aldehyde with indole motivated to investigate the reaction of a dialdehyde and indole to
give novel structures with potential antibacterial activities.
Methods:
Compounds were yielded by chemical synthesis and purified using column chromatography.
The antibacterial activity was determined as minimal inhibitory growth activity in cultures
of Gram-positive strains of Staphylococcus aureus and Enterococcus species.
Results:
Cyclohepta[2,3-b]indoles have been yielded in a one-step reaction procedure with indole
substitutions at the cycloheptane central core matching a solution for achieving fused novel cycloalkane
indoles with functionalized residues of promising biological activity. So far fused cycloalkane
indoles have not been available in a one-step procedure and moreover, core functionalizations
have been additional challenges. Various indole substitutions have been done to provide a
first set of compounds.
Conclusion:
Substituent-dependent effects have been suggested to influence the antibacterial activity
and first compounds were identified with specific Staphylococcus activities and Enterococcus
species effects towards Enterococcus faecalis as critical pathogens in the hospital with upcoming
resistances against standard antibiotics.
Collapse
Affiliation(s)
- Andreas Hilgeroth
- Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle, Germany
| | - Kaveh Yasrebi
- Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle, Germany
| | - Sibel Suzen
- Pharmaceutical Chemistry Department Tandogan, Ankara University, 06100 Ankara, Turkey
| | - Tobias Hertlein
- Institute of Molecular Infection Biology, Julius-Maximilians-University Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzbug, Germany
| | - Knut Ohlsen
- Institute of Molecular Infection Biology, Julius-Maximilians-University Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzbug, Germany
| | - Michael Lalk
- Institute of Biochemistry, Ernst-Moritz-Arndt University Greifswald, Felix-Hausdorff-Str. 4, 17487 Greifswald, Germany
| |
Collapse
|
50
|
Roch M, Lelong E, Panasenko OO, Sierra R, Renzoni A, Kelley WL. Thermosensitive PBP2a requires extracellular folding factors PrsA and HtrA1 for Staphylococcus aureus MRSA β-lactam resistance. Commun Biol 2019; 2:417. [PMID: 31754647 PMCID: PMC6858329 DOI: 10.1038/s42003-019-0667-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/25/2019] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen and represents a clinical challenge because of widespread antibiotic resistance. Methicillin resistant Staphylococcus aureus (MRSA) is particularly problematic and originates by the horizontal acquisition of mecA encoding PBP2a, an extracellular membrane anchored transpeptidase, which confers resistance to β-lactam antibiotics by allosteric gating of its active site channel. Herein, we show that dual disruption of PrsA, a lipoprotein chaperone displaying anti-aggregation activity, together with HtrA1, a membrane anchored chaperone/serine protease, resulted in severe and synergistic attenuation of PBP2a folding that restores sensitivity to β-lactams such as oxacillin. Purified PBP2a has a pronounced unfolding transition initiating at physiological temperatures that leads to irreversible precipitation and complete loss of activity. The concordance of genetic and biochemical data highlights the necessity for extracellular protein folding factors governing MRSA β-lactam resistance. Targeting the PBP2a folding pathway represents a particularly attractive adjuvant strategy to combat antibiotic resistance.
Collapse
Affiliation(s)
- Mélanie Roch
- Department of Microbiology and Molecular Medicine, University Hospital and Medical School of Geneva, 1 rue Michel-Servet, Geneva, CH-1211 Switzerland
| | - Emmanuelle Lelong
- Service of Infectious Diseases and Department of Medical Specialties, University Hospital and Medical School of Geneva, 4 rue Gabrielle-Perret-Gentil, Geneva, CH-1206 Switzerland
| | - Olesya O. Panasenko
- Department of Microbiology and Molecular Medicine, University Hospital and Medical School of Geneva, 1 rue Michel-Servet, Geneva, CH-1211 Switzerland
- Service of Infectious Diseases and Department of Medical Specialties, University Hospital and Medical School of Geneva, 4 rue Gabrielle-Perret-Gentil, Geneva, CH-1206 Switzerland
| | - Roberto Sierra
- Department of Microbiology and Molecular Medicine, University Hospital and Medical School of Geneva, 1 rue Michel-Servet, Geneva, CH-1211 Switzerland
| | - Adriana Renzoni
- Service of Infectious Diseases and Department of Medical Specialties, University Hospital and Medical School of Geneva, 4 rue Gabrielle-Perret-Gentil, Geneva, CH-1206 Switzerland
| | - William L. Kelley
- Department of Microbiology and Molecular Medicine, University Hospital and Medical School of Geneva, 1 rue Michel-Servet, Geneva, CH-1211 Switzerland
| |
Collapse
|