1
|
Shi H, Liu X, Zhao P, Huang W, Wang H, Jin H, Zhu J, Wang J, Li T. Possibility and Potenzial Intervention Targets of Saffron Extract in the Treatment of Atopic Dermatitis: A Review. PLANTA MEDICA 2025; 91:338-352. [PMID: 39947646 DOI: 10.1055/a-2538-5769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Atopic dermatitis (AD) is a chronic, recurrent inflammatory skin disorder characterized by dry skin, eczema-like lesions, and severe itching. The multifaceted etiology of AD, which is not yet fully understood, includes genetic predispositions, immune dysfunctions(such as an impaired skin barrier and abnormal immune regulation), imbalances in the skin microbiota, and environmental factors, among others. In the field of AD treatment, the combination of traditional Chinese medicine and modern medicine is becoming an emerging trend. Given the potenzial side effects and reduced efficacy of conventional therapeutic drugs, Chinese herbal medicines offer patients new treatment options because of their unique efficacy and low toxicity. Some saffron extracts derived from saffron and gardenia, such as crocin, crocetin, and safranal, have shown promising potenzial in the treatment of AD. These natural ingredients not only possess anti-inflammatory and immunomodulatory properties similar to those of traditional Chinese medicines but also demonstrate excellent effects in promoting the repair of damaged skin barriers. Therefore, this article reviews the therapeutic potenzial of saffron extract in the treatment of AD, with a special focus on its mechanisms and potenzial interventions, while emphasizing the importance of herbal medicines as alternatives to traditional treatments, providing AD patients with safer and more effective treatment options.
Collapse
Affiliation(s)
- Huiyang Shi
- Xiangya School of Medicine, Central South University, Changsha, P. R. China
| | - Xuan Liu
- Xiangya School of Public Health, Central South University, Changsha, P. R. China
| | - Peiyi Zhao
- Xiangya School of Medicine, Central South University, Changsha, P. R. China
| | - Wei Huang
- Xiangya School of Medicine, Central South University, Changsha, P. R. China
| | - Hebin Wang
- Xiangya School of Medicine, Central South University, Changsha, P. R. China
| | - Heying Jin
- Xiangya School of Medicine, Central South University, Changsha, P. R. China
| | - Junyou Zhu
- Department of Burn, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Jianwu Wang
- Xiangya School of Public Health, Central South University, Changsha, P. R. China
| | - Tianjiao Li
- Xiangya School of Public Health, Central South University, Changsha, P. R. China
| |
Collapse
|
2
|
Xu Y, Wang L, Guo D, Wang Y, Liu X, Sun Y, Wang R, Sun L, Jiang P, Liu Q, Wang B, Yan M, Zhao Y. Baohuoside I targets SaeR as an antivirulence strategy to disrupt MRSA biofilm formation and pathogenicity. NPJ Biofilms Microbiomes 2025; 11:45. [PMID: 40102466 PMCID: PMC11920273 DOI: 10.1038/s41522-025-00681-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 03/08/2025] [Indexed: 03/20/2025] Open
Abstract
The emergence of methicillin-resistant Staphylococcus aureus (MRSA) represents a critical global health challenge, making the SaeRS two-component system (TCS), a key regulator of S. aureus virulence, an ideal target for novel therapeutic approaches. In this study, virtual screening and thermal shift assays identified Baohuoside I (BI), a flavonol glycoside, as a potent inhibitor of the SaeR response regulator. BI significantly attenuated S. aureus pathogenicity without bactericidal effects, suppressing the expression of key virulence factors, such as hemolysin A (Hla) and Panton-Valentine leukocidin (PVL), while modulating immune evasion pathways. Additionally, BI disrupted biofilm formation, promoting the development of porous, less structured biofilms. Biochemical assays, including EMSA, CETSA, fluorescence quenching, and SPR, confirmed strong binding interactions between SaeR and BI. In vivo, BI demonstrated therapeutic efficacy in Galleria mellonella and rat MRSA models. These findings establish BI as a promising lead for nonbactericidal therapies to combat MRSA infections and mitigate resistance.
Collapse
Affiliation(s)
- Yueshan Xu
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Li Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Dongbin Guo
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yueying Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xinyao Liu
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yun Sun
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Rong Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Luanbiao Sun
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Peitong Jiang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Quan Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun, China
| | - Bingmei Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China.
| | - Ming Yan
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China.
| | - Yicheng Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun, China.
- Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China.
| |
Collapse
|
3
|
Jahangiri A, Dahaghin S, Malekara E, Halabian R, Mahboobi M, Behzadi E, Sedighian H. Highly sensitive detection of Staphylococcus aureus α-hemolysin protein (Hla or α-toxin) by apta-qPCR. J Microbiol Methods 2025; 229:107084. [PMID: 39742925 DOI: 10.1016/j.mimet.2024.107084] [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: 05/07/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Alpha-toxin of Staphylococcus aureus belongs to the pore-forming toxin (PFT) family, which can lyse red and white blood cells. In addition to the existence of the hla gene in the majority of S. aureus strains (about 95 %), higher expression exhibits enhanced pathogenicity to the bacteria. Various methods, such as antibodies and aptamers, could serve to detect this toxin. In the current study, for the first time, an improved sandwich aptamer-antibody-based method was developed using specific murine polyclonal antibodies and a specific aptamer to detect a wide range of α-toxin levels. Denatured recombinant α-toxin was administered to mice to trigger the production of specific antibodies, which were subsequently purified from immune sera. These antibodies served as capturers in the designed apta-qPCR assay, with an aptamer employed as a detector. The results showed that spiked α-toxin in the undiluted serum samples could detect α-toxin between 300 and 0.5 ng/mL with no cross-reactivity. The coefficient of variation (CV) percent of intra- and inter-assays were 0.84 and 1.06, respectively. Since in the apta-qPCR assay, a combination of specific polyclonal antibodies as capture and specific aptamer along with real-time PCR (qPCR) sensitivity is used, this robust method could be used in diagnostic laboratories to detect various levels of the toxin in human serum samples.
Collapse
Affiliation(s)
- Abolfazl Jahangiri
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Samira Dahaghin
- Department of Microbiology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ehsan Malekara
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahdieh Mahboobi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- The Academy of Medical Sciences of Iran, Tehran, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Kim YG, Lee JH, Kim SH, Park SY, Kim YJ, Ryu CM, Seo HW, Lee JT. Inhibition of Biofilm Formation in Cutibacterium acnes, Staphylococcus aureus, and Candida albicans by the Phytopigment Shikonin. Int J Mol Sci 2024; 25:2426. [PMID: 38397101 PMCID: PMC10888572 DOI: 10.3390/ijms25042426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Skin microbiota, such as acne-related Cutibacterium acnes, Staphylococcus aureus, and fungal Candida albicans, can form polymicrobial biofilms with greater antimicrobial tolerance to traditional antimicrobial agents and host immune systems. In this study, the phytopigment shikonin was investigated against single-species and multispecies biofilms under aerobic and anaerobic conditions. Minimum inhibitory concentrations of shikonin were 10 µg/mL against C. acnes, S. aureus, and C. albicans, and at 1-5 µg/mL, shikonin efficiently inhibited single biofilm formation and multispecies biofilm development by these three microbes. Shikonin increased porphyrin production in C. acnes, inhibited cell aggregation and hyphal formation by C. albicans, decreased lipase production, and increased hydrophilicity in S. aureus. In addition, shikonin at 5 or 10 µg/mL repressed the transcription of various biofilm-related genes and virulence-related genes in C. acnes and downregulated the gene expression levels of the quorum-sensing agrA and RNAIII, α-hemolysin hla, and nuclease nuc1 in S. aureus, supporting biofilm inhibition. In addition, shikonin prevented multispecies biofilm development on porcine skin, and the antimicrobial efficacy of shikonin was recapitulated in a mouse infection model, in which it promoted skin regeneration. The study shows that shikonin inhibits multispecies biofilm development by acne-related skin microbes and might be useful for controlling bacterial infections.
Collapse
Affiliation(s)
- Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea; (Y.-G.K.); (J.-H.L.); (S.-H.K.); (S.-Y.P.)
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea; (Y.-G.K.); (J.-H.L.); (S.-H.K.); (S.-Y.P.)
| | - Sang-Hun Kim
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea; (Y.-G.K.); (J.-H.L.); (S.-H.K.); (S.-Y.P.)
| | - Sun-Young Park
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea; (Y.-G.K.); (J.-H.L.); (S.-H.K.); (S.-Y.P.)
| | - Yu-Jeong Kim
- Biosystems & Bioengineering Program, University of Science and Technology (UST), Daejeon Campus, Daejeon 34113, Republic of Korea; (Y.-J.K.); (C.-M.R.)
| | - Choong-Min Ryu
- Biosystems & Bioengineering Program, University of Science and Technology (UST), Daejeon Campus, Daejeon 34113, Republic of Korea; (Y.-J.K.); (C.-M.R.)
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Hwi-Won Seo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jin-Tae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea; (Y.-G.K.); (J.-H.L.); (S.-H.K.); (S.-Y.P.)
| |
Collapse
|
5
|
Jiang F, Chen Y, Yu J, Zhang F, Liu Q, He L, Musha H, Du J, Wang B, Han P, Chen X, Tang J, Li M, Shen H. Repurposed Fenoprofen Targeting SaeR Attenuates Staphylococcus aureus Virulence in Implant-Associated Infections. ACS CENTRAL SCIENCE 2023; 9:1354-1373. [PMID: 37521790 PMCID: PMC10375895 DOI: 10.1021/acscentsci.3c00499] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 08/01/2023]
Abstract
Implant-associated infections (IAIs) caused by S. aureus can result in serious challenges after orthopedic surgery. Due to biofilm formation and antibiotic resistance, this refractory infection is highly prevalent, and finding drugs to attenuate bacterial virulence is becoming a rational alternative strategy. In S. aureus, the SaeRS two-component system (TCS) plays a key role in the production of over 20 virulence factors and the pathogenesis of the bacterium. Here, by conducting a structure-based virtual screening against SaeR, we identified that fenoprofen, a USA Food and Drug Administration (FDA)-approved nonsteroid anti-inflammatory drug (NSAID), had excellent inhibitory potency against the response regulator SaeR protein. We showed that fenoprofen attenuated the virulence of S. aureus without drug resistance. In addition, it was helpful in relieving osteolysis and restoring the walking ability of mice in vitro and in implant-associated infection models. More importantly, fenoprofen treatment suppressed biofilm formation and changed the biofilm structure, which caused S. aureus to form loose and porous biofilms that were more vulnerable to infiltration and elimination by leukocytes. Our results reveal that fenoprofen is a potent antivirulence agent with potential value in clinical applications and that SaeR is a drug target against S. aureus implant-associated infections.
Collapse
Affiliation(s)
- Feng Jiang
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Yingjia Chen
- Drug
Discovery and Design Center, State Key Laboratory of Drug Research,
Shanghai Institute of Materia Medica, Chinese
Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- Department
of Pharmacy, University of Chinese Academy
of Sciences, No.19A Yuan
Road, Beijing 100049, China
| | - Jinlong Yu
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Feiyang Zhang
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Qian Liu
- Department
of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Lei He
- Department
of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Hamushan Musha
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Jiafei Du
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Boyong Wang
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Pei Han
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Xiaohua Chen
- Department
of Infectious Diseases, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Jin Tang
- Department
of Clinical Laboratory, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Min Li
- Department
of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
- Faculty of
Medical Laboratory Science, Shanghai Jiaotong
University School of Medicine, Shanghai 200025, China
| | - Hao Shen
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| |
Collapse
|
6
|
Martínez A, Stashenko EE, Sáez RT, Zafra G, Ortiz C. Effect of Essential Oil from Lippia origanoides on the Transcriptional Expression of Genes Related to Quorum Sensing, Biofilm Formation, and Virulence of Escherichia coli and Staphylococcus aureus. Antibiotics (Basel) 2023; 12:antibiotics12050845. [PMID: 37237748 DOI: 10.3390/antibiotics12050845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Microbial infections resistant to conventional antibiotics constitute one of the most important causes of mortality in the world. In some bacterial species, such as Escherichia coli and Staphylococcus aureus pathogens, biofilm formation can favor their antimicrobial resistance. These biofilm-forming bacteria produce a compact and protective matrix, allowing their adherence and colonization to different surfaces, and contributing to resistance, recurrence, and chronicity of the infections. Therefore, different therapeutic alternatives have been investigated to interrupt both cellular communication routes and biofilm formation. Among these, essential oils (EO) from Lippia origanoides thymol-carvacrol II chemotype (LOTC II) plants have demonstrated biological activity against different biofilm-forming pathogenic bacteria. In this work, we determined the effect of LOTC II EO on the expression of genes associated with quorum sensing (QS) communication, biofilm formation, and virulence of E. coli ATCC 25922 and S. aureus ATCC 29213. This EO was found to have high efficacy against biofilm formation, decreasing-by negative regulation-the expression of genes involved in motility (fimH), adherence and cellular aggregation (csgD), and exopolysaccharide production (pgaC) in E. coli. In addition, this effect was also determined in S. aureus where the L. origanoides EO diminished the expression of genes involved in QS communication (agrA), production of exopolysaccharides by PIA/PNG (icaA), synthesis of alpha hemolysin (hla), transcriptional regulators of the production of extracellular toxins (RNA III), QS and biofilm formation transcriptional regulators (sarA) and global regulators of biofilm formation (rbf and aur). Positive regulation was observed on the expression of genes encoding inhibitors of biofilm formation (e.g., sdiA and ariR). These findings suggest that LOTCII EO can affect biological pathways associated with QS communication, biofilm formation, and virulence of E. coli and S. aureus at subinhibitory concentrations and could be a promising candidate as a natural antibacterial alternative to conventional antibiotics.
Collapse
Affiliation(s)
- Andrés Martínez
- Grupo de Investigación en Bioquímica y Microbiología (GIBIM), Escuela de Microbiología, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga 680002, Colombia
| | - Elena E Stashenko
- Escuela de Química, Centro de Cromatografía y Espectrometría de Masas (CROM-MASS), Universidad Industrial de Santander, Bucaramanga 680002, Colombia
| | - Rodrigo Torres Sáez
- Grupo de Investigación en Bioquímica y Microbiología (GIBIM), Escuela de Microbiología, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga 680002, Colombia
| | - German Zafra
- Grupo de Investigación en Bioquímica y Microbiología (GIBIM), Escuela de Microbiología, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga 680002, Colombia
| | - Claudia Ortiz
- Grupo de Investigación en Bioquímica y Microbiología (GIBIM), Escuela de Microbiología, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga 680002, Colombia
| |
Collapse
|
7
|
Figueiredo CSSES, de Oliveira PV, Saminez WFDS, Diniz RM, Mendonça JSP, Silva LDS, Paiva MYM, do Nascimento MDS, Aliança ASDS, Zagmignan A, Rodrigues JFS, Souza JCDS, Grisotto MAG, da Silva LCN. Immunomodulatory Effects of Cinnamaldehyde in Staphylococcus aureus-Infected Wounds. Molecules 2023; 28:molecules28031204. [PMID: 36770872 PMCID: PMC9921375 DOI: 10.3390/molecules28031204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/12/2022] [Accepted: 12/23/2022] [Indexed: 01/28/2023] Open
Abstract
Cinnamaldehyde (CNM) is an essential-oil component with reported anti-infective, anti-inflammatory, and healing effects, making it an interesting compound for the treatment of wound infection. Herein, we evaluated the effects of topical administration of CNM in experimental wounds infected by Staphylococcus aureus. Swiss mice (n = 12/group) were randomly allocated into three groups (CON: animals with uninfected lesions; Sa: animals with untreated infected lesions; Sa + CNM: animals with infected wounds and treated with CNM). Excisional lesions (64 mm2) were induced at the dorsal area followed by the addition of S. aureus (80 μL of a 1.5 × 108 CFU/mL bacterial suspension). The wounds were treated with CNM (200 μg/wound/day) or vehicle (2% DMSO) for 10 days. Skin samples were taken on the 3rd or 10th treatment day for quantification of inflammatory mediators, bacterial load, immunophenotyping, and histological analysis. The treatment with CNM improved the healing process and attenuated the severity of skin lesions infected by S. aureus. These effects were associated with significant decreases in bacterial loads in CNM-treated wounds. The levels of neutrophils, TNF-α, IL-6, NO, and VEGF were decreased in the lesions treated with CNM. Taken together, these data provide further evidence of the effectiveness of CNM for the treatment of skin infections.
Collapse
Affiliation(s)
- Cristiane Santos Silva e Silva Figueiredo
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil
- Rede de Biodiversidade e Biotecnologia da Amazônia Legal (BIONORTE), São Luís 65075-120, Brazil
| | | | | | - Roseana Muniz Diniz
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil
| | | | - Lucas dos Santos Silva
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil
| | | | | | | | - Adrielle Zagmignan
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil
| | | | | | | | - Luís Cláudio Nascimento da Silva
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil
- Rede de Biodiversidade e Biotecnologia da Amazônia Legal (BIONORTE), São Luís 65075-120, Brazil
- Correspondence:
| |
Collapse
|
8
|
Kim J, Kim BE, Berdyshev E, Bronova I, Bin L, Bae J, Kim S, Kim HY, Lee UH, Kim MS, Kim H, Lee J, Hall CF, Hui-Beckman J, Chang Y, Bronoff AS, Hwang D, Lee HY, Goleva E, Ahn K, Leung DYM. Staphylococcus aureus causes aberrant epidermal lipid composition and skin barrier dysfunction. Allergy 2023; 78:1292-1306. [PMID: 36609802 DOI: 10.1111/all.15640] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Staphylococcus (S) aureus colonization is known to cause skin barrier disruption in atopic dermatitis (AD) patients. However, it has not been studied how S. aureus induces aberrant epidermal lipid composition and skin barrier dysfunction. METHODS Skin tape strips (STS) and swabs were obtained from 24 children with AD (6.0 ± 4.4 years) and 16 healthy children (7.0 ± 4.5 years). Lipidomic analysis of STS samples was performed by mass spectrometry. Skin levels of methicillin-sensitive and methicillin-resistant S. aureus (MSSA and MRSA) were evaluated. The effects of MSSA and MRSA were evaluated in primary human keratinocytes (HEKs) and organotypic skin cultures. RESULTS AD and organotypic skin colonized with MRSA significantly increased the proportion of lipid species with nonhydroxy fatty acid sphingosine ceramide with palmitic acid ([N-16:0 NS-CER], sphingomyelins [16:0-18:0 SM]), and lysophosphatidylcholines [16:0-18:0 LPC], but significantly reduced the proportion of corresponding very long-chain fatty acids (VLCFAs) species (C22-28) compared to the skin without S. aureus colonization. Significantly increased transepidermal water loss (TEWL) was found in MRSA-colonized AD skin. S. aureus indirectly through interleukin (IL)-1β, tumor necrosis factor (TNF)-α, IL-6, and IL-33 inhibited expression of fatty acid elongase enzymes (ELOVL3 and ELOVL4) in HEKs. ELOVL inhibition was more pronounced by MRSA and resulted in TEWL increase in organotypic skin. CONCLUSION Aberrant skin lipid profiles and barrier dysfunction are associated with S. aureus colonization in AD patients. These effects are attributed to the inhibition of ELOVLs by S. aureus-induced IL-1β, TNF-α, IL-6, and IL-33 seen in keratinocyte models and are more prominent in MRSA than MSSA.
Collapse
Affiliation(s)
- Jihyun Kim
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Byung Eui Kim
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Evgeny Berdyshev
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Irina Bronova
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Lianghua Bin
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Jaewoong Bae
- R&D Institute, BioEleven Co., Ltd., Seoul, Korea
| | - Seokjin Kim
- R&D Institute, BioEleven Co., Ltd., Seoul, Korea
| | - Hye-Young Kim
- Department of Pediatrics, Medical Research Institute of Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Un Ha Lee
- Department of Dermatology, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Myoung Shin Kim
- Department of Dermatology, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Hyunmi Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jinyoung Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Clifton F Hall
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | | | - Yunhee Chang
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju, Korea
| | | | - Dasom Hwang
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju, Korea
| | - Hae-Young Lee
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju, Korea
| | - Elena Goleva
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| |
Collapse
|
9
|
Jean-Pierre V, Boudet A, Sorlin P, Menetrey Q, Chiron R, Lavigne JP, Marchandin H. Biofilm Formation by Staphylococcus aureus in the Specific Context of Cystic Fibrosis. Int J Mol Sci 2022; 24:ijms24010597. [PMID: 36614040 PMCID: PMC9820612 DOI: 10.3390/ijms24010597] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen whose characteristics support its success in various clinical settings including Cystic Fibrosis (CF). In CF, S. aureus is indeed the most commonly identified opportunistic pathogen in children and the overall population. S. aureus colonization/infection, either by methicillin-susceptible or methicillin-resistant strains, will become chronic in about one third of CF patients. The persistence of S. aureus in CF patients' lungs, despite various eradication strategies, is favored by several traits in both host and pathogen. Among the latter, living in biofilm is a highly protective way to survive despite deleterious environmental conditions, and is a common characteristic shared by the main pathogens identified in CF. This is why CF has earned the status of a biofilm-associated disease for several years now. Biofilm formation by S. aureus, and the molecular mechanisms governing and regulating it, have been extensively studied but have received less attention in the specific context of CF lungs. Here, we review the current knowledge on S. aureus biofilm in this very context, i.e., the importance, study methods, molecular data published on mono- and multi-species biofilm and anti-biofilm strategies. This focus on studies including clinical isolates from CF patients shows that they are still under-represented in the literature compared with studies based on reference strains, and underlines the need for such studies. Indeed, CF clinical strains display specific characteristics that may not be extrapolated from results obtained on laboratory strains.
Collapse
Affiliation(s)
- Vincent Jean-Pierre
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 34093 Montpellier, France
| | - Agathe Boudet
- VBIC—Virulence Bactérienne et Infections Chroniques, Université de Montpellier, INSERM U1047, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30900 Nîmes, France
| | - Pauline Sorlin
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, 34093 Montpellier, France
| | - Quentin Menetrey
- INFINITE—Institute for Translational Research in Inflammation, Université de Lille, INSERM U1286, CHU Lille, 59000 Lille, France
| | - Raphaël Chiron
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, Centre de Ressources et de Compétences de la Mucoviscidose, CHU Montpellier, 34295 Montpellier, France
| | - Jean-Philippe Lavigne
- VBIC—Virulence Bactérienne et Infections Chroniques, Université de Montpellier, INSERM U1047, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30900 Nîmes, France
| | - Hélène Marchandin
- HSM—HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 34093 Montpellier, France
- Correspondence:
| |
Collapse
|
10
|
In Silico Genome-Scale Analysis of Molecular Mechanisms Contributing to the Development of a Persistent Infection with Methicillin-Resistant Staphylococcus aureus (MRSA) ST239. Int J Mol Sci 2022; 23:ijms232416086. [PMID: 36555727 PMCID: PMC9781258 DOI: 10.3390/ijms232416086] [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: 10/30/2022] [Revised: 12/05/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
The increasing frequency of isolation of methicillin-resistant Staphylococcus aureus (MRSA) limits the chances for the effective antibacterial therapy of staphylococcal diseases and results in the development of persistent infection such as bacteremia and osteomyelitis. The aim of this study was to identify features of the MRSAST239 0943-1505-2016 (SA943) genome that contribute to the formation of both acute and chronic musculoskeletal infections. The analysis was performed using comparative genomics data of the dominant epidemic S. aureus lineages, namely ST1, ST8, ST30, ST36, and ST239. The SA943 genome encodes proteins that provide resistance to the host's immune system, suppress immunological memory, and form biofilms. The molecular mechanisms of adaptation responsible for the development of persistent infection were as follows: amino acid substitution in PBP2 and PBP2a, providing resistance to ceftaroline; loss of a large part of prophage DNA and restoration of the nucleotide sequence of beta-hemolysin, that greatly facilitates the escape of phagocytosed bacteria from the phagosome and formation of biofilms; dysfunction of the AgrA system due to the presence of psm-mec and several amino acid substitutions in the AgrC; partial deletion of the nucleotide sequence in genomic island vSAβ resulting in the loss of two proteases of Spl-operon; and deletion of SD repeats in the SdrE amino acid sequence.
Collapse
|
11
|
Han J, Poma A. Molecular Targets for Antibody-Based Anti-Biofilm Therapy in Infective Endocarditis. Polymers (Basel) 2022; 14:3198. [PMID: 35956712 PMCID: PMC9370930 DOI: 10.3390/polym14153198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Infective endocarditis (IE) is a heart disease caused by the infection of heart valves, majorly caused by Staphilococcus aureus. IE is initiated by bacteria entering the blood circulation in favouring conditions (e.g., during invasive procedures). So far, the conventional antimicrobial strategies based on the usage of antibiotics remain the major intervention for treating IE. Nevertheless, the therapeutic efficacy of antibiotics in IE is limited not only by the bacterial drug resistance, but also by the formation of biofilms, which resist the penetration of antibiotics into bacterial cells. To overcome these drawbacks, the development of anti-biofilm treatments that can expose bacteria and make them more susceptible to the action of antibiotics, therefore resulting in reduced antimicrobial resistance, is urgently required. A series of anti-biofilm strategies have been developed, and this review will focus in particular on the development of anti-biofilm antibodies. Based on the results previously reported in the literature, several potential anti-biofilm targets are discussed, such as bacterial adhesins, biofilm matrix and bacterial toxins, covering their antigenic properties (with the identification of potential promising epitopes), functional mechanisms, as well as the antibodies already developed against these targets and, where feasible, their clinical translation.
Collapse
Affiliation(s)
- Jiahe Han
- UCL Institute of Cardiovascular Science, The Rayne Building, 5 University Street, London WC1E 6JF, UK
| | - Alessandro Poma
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, Royal Free Hospital, UCL Medical School, Rowland Hill Street, London NW3 2PF, UK
| |
Collapse
|
12
|
Bisdemethoxycurcumin Reduces Methicillin-Resistant Staphylococcus aureus Expression of Virulence-Related Exoproteins and Inhibits the Biofilm Formation. Toxins (Basel) 2021; 13:toxins13110804. [PMID: 34822588 PMCID: PMC8625963 DOI: 10.3390/toxins13110804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/11/2021] [Accepted: 11/13/2021] [Indexed: 12/18/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a major pathogen of nosocomial infection, which is resistant to most antibiotics. Presently, anti-virulence therapy and anti-biofilm therapy are considered to be promising alternatives. In the current work, we investigated the influence of bisdemethoxycurcumin (BDMC) on the virulence-related exoproteins and the biofilm formation using a reference strain and clinic isolated strains. Western blotting, quantitative RT-PCR, and tumor necrosis factor (TNF) release assay were performed to assess the efficacy of BDMC in reducing the expression of Staphylococcus enterotoxin-related exoproteins (enterotoxin A, enterotoxin B) and α-toxin in MRSA. The anti-biofilm activity of BDMC was evaluated through a biofilm inhibition assay. The study suggests that sub-inhibitory concentrations of BDMC significantly inhibited the expression of sea, seb, and hla at the mRNA level in MRSA. Moreover, the expression of virulence-related exoproteins was significantly decreased by down-regulating accessory gene regulator agr, and the inhibition of biofilms formation was demonstrated by BDMC at sub-inhibitory concentrations. Consequently, the study suggests that BDMC may be a potential natural antibacterial agent to release the pressure brought by antibiotic resistance.
Collapse
|
13
|
Zheng X, Chen L, Zeng W, Liao W, Wang Z, Tian X, Fang R, Sun Y, Zhou T. Antibacterial and Anti-biofilm Efficacy of Chinese Dragon's Blood Against Staphylococcus aureus Isolated From Infected Wounds. Front Microbiol 2021; 12:672943. [PMID: 34149659 PMCID: PMC8213214 DOI: 10.3389/fmicb.2021.672943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/07/2021] [Indexed: 01/01/2023] Open
Abstract
Chinese dragon’s blood (CDB), a characteristic red resin, is an important traditional Chinese medicine (TCM), and empiric therapy of infected wounds with CDB is performed in clinical settings. For the first time, we herein report the antibacterial and anti-biofilm efficacy of CDB against Staphylococcus aureus (S. aureus). Antimicrobial susceptibility testing, growth curve assay, time-kill curve assay, crystal violet biofilm assay, scanning electron microscope (SEM) analysis, cell membrane tests, and quantitative real-time polymerase chain reaction (qRT-PCR) were used for this purpose. The results suggested that the minimum inhibitory concentration (MIC) values of CDB against S. aureus ranged from 32 to 128 μg/mL. Growth curves and time-kill curves confirmed that CDB could inhibit the growth of S. aureus. The biofilm formation ability and the expression levels of saeR, saeS, and hla of S. aureus in the presence and absence of CDB were statistically significant (P < 0.01). The results of SEM analysis and cell membrane tests revealed that exposure to CDB had some destructive effects on S. aureus cells. In conclusion, CDB exhibits positive antibacterial activity against S. aureus. Moreover, CDB could reduce the biofilm formation and the virulence factors of S. aureus by downregulating the expression levels of saeR, saeS, and hla genes. These findings indicated that CDB has immense potential to serve as a viable alternative for the treatment of infected wounds caused by S. aureus in clinical settings.
Collapse
Affiliation(s)
- Xiangkuo Zheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lijiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weiliang Zeng
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenli Liao
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongyong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuebin Tian
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Renchi Fang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yao Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
Antibiofilm Activity of Phorbaketals from the Marine Sponge Phorbas sp. against Staphylococcus aureus. Mar Drugs 2021; 19:md19060301. [PMID: 34073814 PMCID: PMC8225198 DOI: 10.3390/md19060301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 01/02/2023] Open
Abstract
Biofilm formation by Staphylococcus aureus plays a critical role in the persistence of chronic infections due to its tolerance against antimicrobial agents. Here, we investigated the antibiofilm efficacy of six phorbaketals: phorbaketal A (1), phorbaketal A acetate (2), phorbaketal B (3), phorbaketal B acetate (4), phorbaketal C (5), and phorbaketal C acetate (6), isolated from the Korean marine sponge Phorbas sp. Of these six compounds, 3 and 5 were found to be effective inhibitors of biofilm formation by two S. aureus strains, which included a methicillin-resistant S. aureus. In addition, 3 also inhibited the production of staphyloxanthin, which protects microbes from reactive oxygen species generated by neutrophils and macrophages. Transcriptional analyses showed that 3 and 5 inhibited the expression of the biofilm-related hemolysin gene hla and the nuclease gene nuc1.
Collapse
|
15
|
Monoclonal Antibodies Targeting Surface-Exposed and Secreted Proteins from Staphylococci. Vaccines (Basel) 2021; 9:vaccines9050459. [PMID: 34064471 PMCID: PMC8147999 DOI: 10.3390/vaccines9050459] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/14/2021] [Accepted: 04/30/2021] [Indexed: 02/01/2023] Open
Abstract
Staphylococci (specifically Staphylococcus aureus and Staphylococcus epidermidis) are the causative agents of diseases ranging from superficial skin and soft tissue infections to severe conditions such as fatal pneumonia, bacteremia, sepsis and endocarditis. The widespread and indiscriminate use of antibiotics has led to serious problems of resistance to staphylococcal disease and has generated a renewed interest in alternative therapeutic agents such as vaccines and antibodies. Staphylococci express a large repertoire of surface and secreted virulence factors, which provide mechanisms (adhesion, invasion and biofilm development among others) for both bacterial survival in the host and evasion from innate and adaptive immunity. Consequently, the development of antibodies that target specific antigens would provide an effective protective strategy against staphylococcal infections. In this review, we report an update on efforts to develop anti-staphylococci monoclonal antibodies (and their derivatives: minibodies, antibody–antibiotic conjugates) and the mechanism by which such antibodies can help fight infections. We also provide an overview of mAbs used in clinical trials and highlight their therapeutic potential in various infectious contexts.
Collapse
|
16
|
Karauzum H, Venkatasubramaniam A, Adhikari RP, Kort T, Holtsberg FW, Mukherjee I, Mednikov M, Ortines R, Nguyen NTQ, Doan TMN, Diep BA, Lee JC, Aman MJ. IBT-V02: A Multicomponent Toxoid Vaccine Protects Against Primary and Secondary Skin Infections Caused by Staphylococcus aureus. Front Immunol 2021; 12:624310. [PMID: 33777005 PMCID: PMC7987673 DOI: 10.3389/fimmu.2021.624310] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus causes a wide range of diseases from skin infections to life threatening invasive diseases such as bacteremia, endocarditis, pneumonia, surgical site infections, and osteomyelitis. Skin infections such as furuncles, carbuncles, folliculitis, erysipelas, and cellulitis constitute a large majority of infections caused by S. aureus (SA). These infections cause significant morbidity, healthcare costs, and represent a breeding ground for antimicrobial resistance. Furthermore, skin infection with SA is a major risk factor for invasive disease. Here we describe the pre-clinical efficacy of a multicomponent toxoid vaccine (IBT-V02) for prevention of S. aureus acute skin infections and recurrence. IBT-V02 targets six SA toxins including the pore-forming toxins alpha hemolysin (Hla), Panton-Valentine leukocidin (PVL), leukocidin AB (LukAB), and the superantigens toxic shock syndrome toxin-1 and staphylococcal enterotoxins A and B. Immunization of mice and rabbits with IBT-V02 generated antibodies with strong neutralizing activity against toxins included in the vaccine, as well as cross-neutralizing activity against multiple related toxins, and protected against skin infections by several clinically relevant SA strains of USA100, USA300, and USA1000 clones. Efficacy of the vaccine was also shown in non-naïve mice pre-exposed to S. aureus. Furthermore, vaccination with IBT-V02 not only protected mice from a primary infection but also demonstrated lasting efficacy against a secondary infection, while prior challenge with the bacteria alone was unable to protect against recurrence. Serum transfer studies in a primary infection model showed that antibodies are primarily responsible for the protective response.
Collapse
Affiliation(s)
| | | | | | - Tom Kort
- Integrated BioTherapeutics, Rockville, MD, United States
| | | | | | - Mark Mednikov
- Integrated BioTherapeutics, Rockville, MD, United States
| | - Roger Ortines
- Integrated BioTherapeutics, Rockville, MD, United States
| | - Nhu T. Q. Nguyen
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Thien M. N. Doan
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Binh An Diep
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jean C. Lee
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - M. Javad Aman
- Integrated BioTherapeutics, Rockville, MD, United States
| |
Collapse
|
17
|
Brandenburg KS, Weaver AJ, Karna SLR, Leung KP. The impact of simultaneous inoculation of Pseudomonas aeruginosa, Staphylococcus aureus, and Candida albicans on rodent burn wounds. Burns 2021; 47:1818-1832. [PMID: 33771422 DOI: 10.1016/j.burns.2021.02.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/13/2021] [Accepted: 02/18/2021] [Indexed: 12/27/2022]
Abstract
Burn wound infection often involves a diverse combination of bacterial and fungal pathogens. In this study, we characterize the mixed species burn wound infection by inoculating the burn surface with 1 × 103/4/5 CFU of Pseudomonas aeruginosa, Staphylococcus aureus, and Candida albicans in a 1:1:1 ratio. Using the revised Walker-Mason scald burn rat model, 168 male Sprague-Dawley rats (350-450 g) subject to ∼10% TBSA burn injury, with or without inoculation, were evaluated for 11 days after burn. In the wound, P. aeruginosa and S. aureus formed robust biofilms as determined by the bacterial tissue load, ∼1 × 109 CFU/g, and expression of key biofilm genes. Interestingly, within 3 days C. albicans achieved tissue loads of ∼1 × 106 CFU/g, but its numbers were significantly reduced beyond the limit of detection in the burn wound by day 7 in partial-thickness injuries and by day 11 in full-thickness injuries. The pathogenic biofilms contributed to burn depth progression, increased release of HMGB-1 into circulation from injured tissue, and significantly elevated the numbers of circulating innate immune cells (Neutrophils, Monocytes, and Basophils). This robust model of multi-species burn wound infection will serve as the basis for the development of new antimicrobials for combating biofilm-based wound infections.
Collapse
Affiliation(s)
- Kenneth S Brandenburg
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX 78234, USA.
| | - Alan J Weaver
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX 78234, USA.
| | - S L Rajasekhar Karna
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX 78234, USA.
| | - Kai P Leung
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JBSA Fort Sam Houston, TX 78234, USA.
| |
Collapse
|
18
|
Saeed K, Sendi P, Arnold WV, Bauer TW, Coraça-Huber DC, Chen AF, Choe H, Daiss JL, Ghert M, Hickok NJ, Nishitani K, Springer BD, Stoodley P, Sculco TP, Brause BD, Parvizi J, McLaren AC, Schwarz EM. Bacterial toxins in musculoskeletal infections. J Orthop Res 2021; 39:240-250. [PMID: 32255540 PMCID: PMC7541548 DOI: 10.1002/jor.24683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/27/2020] [Accepted: 04/01/2020] [Indexed: 02/04/2023]
Abstract
Musculoskeletal infections (MSKIs) remain a major health burden in orthopaedics. Bacterial toxins are foundational to pathogenesis in MSKI, but poorly understood by the community of providers that care for patients with MSKI, inducing an international group of microbiologists, infectious diseases specialists, orthopaedic surgeons and biofilm scientists to review the literature in this field to identify key topics and compile the current knowledge on the role of toxins in MSKI, with the goal of illuminating potential impact on biofilm formation and dispersal as well as therapeutic strategies. The group concluded that further research is needed to maximize our understanding of the effect of toxins on MSKIs, including: (i) further research to identify the roles of bacterial toxins in MSKIs, (ii) establish the understanding of the importance of environmental and host factors and in vivo expression of toxins throughout the course of an infection, (iii) establish the principles of drug-ability of antitoxins as antimicrobial agents in MSKIs, (iv) have well-defined metrics of success for antitoxins as antiinfective drugs, (v) design a cocktail of antitoxins against specific pathogens to (a) inhibit biofilm formation and (b) inhibit toxin release. The applicability of antitoxins as potential antimicrobials in the era of rising antibiotic resistance could meet the needs of day-to-day clinicians.
Collapse
Affiliation(s)
- Kordo Saeed
- University Hospital Southampton NHS Foundation Trust, Department of Microbiology, Microbiology Innovation and Research Unit (MIRU), Southampton, UK; and University of Southampton, School of Medicine, Southampton UK
| | - Parham Sendi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology/ Department of Orthopaedics and Traumatology, University Hospital Basel, University Basel, Basel, Switzerland
| | - William V. Arnold
- Department of Orthopaedic Surgery, Rothman Institute at Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Thomas W. Bauer
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Hospital for Special Surgery, New York, NY, USA
| | - Débora C. Coraça-Huber
- Research Laboratory for Implant Associated Infections (Biofilm Lab), Experimental Orthopaedics, Department of Orthopaedic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Antonia F. Chen
- Department of Orthopaedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Hyonmin Choe
- Department of Orthopaedic Surgery, Yokohama City University, Yokohama, Kanagawa, Japan
| | - John L. Daiss
- Center for Musculoskeletal Research, School of Medicine and Dentistry University of Rochester, Rochester, NY, USA
| | - Michelle Ghert
- Department of Surgery, Division of Orthopaedic Surgery, McMaster University, Hamilton, ON, Canada
| | - Noreen J. Hickok
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA, USA
| | - Kohei Nishitani
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| | - Bryan D. Springer
- OrthoCarolina Hip and Knee Center, Atrium Musculoskeletal Institute, Charlotte, NC, USA
| | - Paul Stoodley
- Departments of Microbial Infection and Immunity and OrthopedicsInfectious Diseases Institute, The Ohio State University, 716 Biomedical Research Tower, 460 West 12th Avenue, Columbus OH, Canada
- National Centre for Microbial Tribology at Southampton (nCATS), National Biofilm Innovation Centre (NBIC), Mechanical Engineering, University of Southampton, Southampton, UK.
| | - Thomas P. Sculco
- Department of Orthopaedic Surgery, Weill Cornell Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Barry D. Brause
- Department of Infectious Diseases, Weill Cornell Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Javad Parvizi
- Department of Orthopaedics, Rothman Institute at Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Alex C. McLaren
- Department of Orthopaedic Surgery, University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester, Rochester, NY, USA
| |
Collapse
|
19
|
Yoon DJ, Nguyen C, Bagood MD, Fregoso DR, Yang HY, Medina Lopez AI, Crawford RW, Tran J, Isseroff RR. Topical Fluoxetine as a Potential Nonantibiotic Adjunctive Therapy for Infected Wounds. J Invest Dermatol 2020; 141:1608-1612.e3. [PMID: 33338500 DOI: 10.1016/j.jid.2020.11.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/13/2020] [Accepted: 11/10/2020] [Indexed: 01/15/2023]
Affiliation(s)
- Daniel J Yoon
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Chuong Nguyen
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Michelle D Bagood
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Daniel R Fregoso
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Hsin-Ya Yang
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Andrea I Medina Lopez
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Robert W Crawford
- Department of Biological Sciences, California State University, Sacramento, Sacramento, California, USA
| | - Jennifer Tran
- Department of Biological Sciences, California State University, Sacramento, Sacramento, California, USA
| | - R Rivkah Isseroff
- Department of Dermatology, University of California, Davis, Sacramento, California, USA.
| |
Collapse
|
20
|
Antibiofilm effects of N,O-acetals derived from 2-amino-1,4-naphthoquinone are associated with downregulation of important global virulence regulators in methicillin-resistant Staphylococcus aureus. Sci Rep 2020; 10:19631. [PMID: 33184312 PMCID: PMC7661526 DOI: 10.1038/s41598-020-76372-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the existing antibiotics, antimicrobial resistance is a major challenge. Consequently, the development of new drugs remains in great demand. Quinones is part of a broad group of molecules that present antibacterial activity besides other biological properties. The main purpose of this study was to evaluate the antibiofilm activities of synthetic N,O-acetals derived from 2-amino-1,4-naphthoquinone [7a: 2-(methoxymethyl)-amino-1,4-naphthoquinone; 7b: 2-(ethoxymethyl)-amino-1,4-naphthoquinone; and 7c: 2-(propynyloxymethyl)-amino-1,4-naphthoquinone] against methicillin-resistant Staphylococcus aureus (MRSA). The derivatives 7b and 7c, specially 7b, caused strong impact on biofilm accumulation. This inhibition was linked to decreased expression of the genes fnbA, spa, hla and psmα3. More importantly, this downregulation was paralleled by the modulation of global virulence regulators. The substitution of 2-ethoxymethyl (7b) in comparison with 2-propynyloxymethyl (7c) enhanced sarA-agr inhibition, decreased fnbA transcripts (positively regulated by sarA) and strongly impaired biofilm accumulation. Indeed, 7b triggered intensive autolysis and was able to eliminate vancomycin-persistent cells. Consequently, 7b is a promising molecule displaying not only antimicrobial effects, but also antibiofilm and antipersistence activities. Therefore, 7b is a good candidate for further studies involving the development of novel and more rational antimicrobials able to act in chronic and recalcitrant infections, associated with biofilm formation.
Collapse
|
21
|
Qian Y, Xia L, Wei L, Li D, Jiang W. Artesunate inhibits Staphylococcus aureus biofilm formation by reducing alpha-toxin synthesis. Arch Microbiol 2020; 203:707-717. [PMID: 33040179 DOI: 10.1007/s00203-020-02077-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/13/2020] [Accepted: 10/01/2020] [Indexed: 11/30/2022]
Abstract
Staphylococcus aureus is one of the most common pathogens in bacterial biofilm infections. Antibiotic treatment for infection caused by S. aureus biofilms is challenging, and few effective strategies have been developed to combat these infections. The aim of this study was to investigate the effect and possible mechanisms of artesunate on the biofilm formation of S. aureus. Bacterial growth curves were determined by a microtiter plate. Biofilm formation was determined by the crystal violet staining method and confocal laser scanning microscopy. Bacterial adhesion was assayed by the colony-counting method. The expression of virulence and adhesion genes was determined by real-time PCR. The hemolytic activity and expression of ɑ-hemolysin were analyzed using rabbit erythrocytes and Western blotting. The results showed that artesunate could significantly inhibit the biofilm formation of S. aureus in a dose-dependent manner. Artesunate could also inhibit bacterial adhesion and the expression of hla, RNAIII and agrA as well as ɑ-hemolysin production. The effect of artesunate on adhesion genes (clfA, clfB, fnbA, fnbB) had strain specificity, but it did not affect the expression of ica genes. The results indicated that artesunate might inhibit ɑ-hemolysin synthesis by the agr system, which inhibits biofilm formation.
Collapse
Affiliation(s)
- Yan Qian
- Department of Pharmacy, The Second Affiliated Hospital, Chongqing Medical University, No. 76, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Li Xia
- Department of Liver Disease, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Lai Wei
- Department of Pharmacy, The Second Affiliated Hospital, Chongqing Medical University, No. 76, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Di Li
- Department of Pharmacy, The Second Affiliated Hospital, Chongqing Medical University, No. 76, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Weiwei Jiang
- Department of Pharmacy, The Second Affiliated Hospital, Chongqing Medical University, No. 76, Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
22
|
Wang T, Zhang P, Lv H, Deng X, Wang J. A Natural Dietary Flavone Myricetin as an α-Hemolysin Inhibitor for Controlling Staphylococcus aureus Infection. Front Cell Infect Microbiol 2020; 10:330. [PMID: 32793508 PMCID: PMC7390952 DOI: 10.3389/fcimb.2020.00330] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus, an important agent for lethal bacterial infections, can cause a broad spectrum of diseases in various host species. The emergence of multidrug-resistant and highly virulent strains has raised increasing concerns about the novel therapeutic strategies or agents available for treating S. aureus infection. The critical role of Hla, an essential virulence determinant, in the pathogenicity of S. aureus renders this toxin an attractive target for effective therapeutic applications. Here, we have identified myricetin as an effective inhibitor of Hla that simultaneously inhibits Hla production and neutralizes Hla activity without affecting bacterial growth. Myricetin treatment reduced the oligomerization of Hla and Hla-mediated biofilm formation. The addition of myricetin to the coinfection system of host cells and S. aureus significantly decreased cell injury and downregulated the inflammatory response in cells. Furthermore, S. aureus-infected mice that received myricetin showed alleviated tissue damage in the lung. Our results indicated that myricetin inhibits S. aureus virulence by targeting Hla and downregulates the inflammatory response in host cells. Overall, in addition to traditional antibiotics with antibacterial activity, myricetin may represent a potential candidate, and strategy for S. aureus infection.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Peng Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hongfa Lv
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuming Deng
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianfeng Wang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
23
|
Forson AM, van der Mei HC, Sjollema J. Impact of solid surface hydrophobicity and micrococcal nuclease production on Staphylococcus aureus Newman biofilms. Sci Rep 2020; 10:12093. [PMID: 32694559 PMCID: PMC7374737 DOI: 10.1038/s41598-020-69084-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is commonly associated with biofilm-related infections and contributes to the large financial loss that accompany nosocomial infections. The micrococcal nuclease Nuc1 enzyme limits biofilm formation via cleavage of eDNA, a structural component of the biofilm matrix. Solid surface hydrophobicity influences bacterial adhesion forces and may as well influence eDNA production. Therefore, it is hypothesized that the impact of Nuc1 activity is dependent on surface characteristics of solid surfaces. For this reason, this study investigated the influence of solid surface hydrophobicity on S. aureus Newman biofilms where Nuc1 is constitutively produced. To this end, biofilms of both a wild-type and a nuc1 knockout mutant strain, grown on glass, salinized glass and Pluronic F-127-coated silanized glass were analysed. Results indicated that biofilms can grow in the presence of Nuc1 activity. Also, Nuc1 and solid surface hydrophobicity significantly affected the biofilm 3D-architecture. In particular, biofilm densities of the wild-type strain on hydrophilic surfaces appeared higher than of the mutant nuc1 knockout strain. Since virulence is related to bacterial cell densities, this suggests that the virulence of S. aureus Newman biofilms is increased by its nuclease production in particular on a hydrophilic surface.
Collapse
Affiliation(s)
- Abigail M Forson
- University of Groningen, University Medical Centre Groningen, Department of Biomedical Engineering, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Henny C van der Mei
- University of Groningen, University Medical Centre Groningen, Department of Biomedical Engineering, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Jelmer Sjollema
- University of Groningen, University Medical Centre Groningen, Department of Biomedical Engineering, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
24
|
Khasawneh AI, Himsawi N, Abu-Raideh J, Salameh MA, Al-Tamimi M, Al Haj Mahmoud S, Saleh T. Status of Biofilm-Forming Genes among Jordanian Nasal Carriers of Methicillin-Sensitive and Methicillin-Resistant Staphylococcus aureus. IRANIAN BIOMEDICAL JOURNAL 2020; 24:386-98. [PMID: 32660224 PMCID: PMC7601545 DOI: 10.29252/ibj.24.6.381] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Biofilm formation in Staphylococcusaureus is a major virulence factor. Both MSSA and MRSA are common causes of community- and hospital-acquired infections and are associated with biofilm formation. The status of biofilm-forming genes has not been explored in Jordanian nasal carriers of S. aureus. This study investigates antibiotic resistance patterns and the prevalence of biofilm-forming genes between MSSA and MRSA in two distinct populations in Jordan. Methods: A total of 35 MSSA and 22 MRSA isolates were recovered from hospitalized patients and medical students at Prince Hamzah Hospital, Jordan. Antibiotic susceptibility was tested using disk diffusion method and Vitek 2 system. The phenotypic biofilm formation was tested using CRA and microtiter plate assays. The prevalence of the biofilm-forming genes was determined using multiplex PCR. Results: Among 57 S.aureus isolates, 22 (38.6%) isolates were MRSA and were highly resistant against benzylpenicillin, oxacillin, and imipenem. The frequencies of the icaADBC were 77.1%, 97.1%, 94.3%, and 97.1% respectively in MSSA compared to 86.4%, 100%, 100%, and 100% in MRSA isolates. On the other hand, the frequency of the fnbA, fnbB, clfA, fib, clfB, ebps, eno, and cna genes was 81.8%, 90.9%, 95.5%, 90.9%, 86.4%, 100%, 100%, and 40.9%, respectively in the MRSA isolates. Conclusion: In both groups, MRSA isolates, in comparison to MSSA, were significantly more resistant to cefoxitin, oxacillin, imipenem, tetracycline, clindamycin, and trimethoprim-sulfamethoxazole. Unexpectedly, biofilm formation and gene prevalence between MRSA and MSSA isolates showed no significant difference, suggesting other potential virulence mechanisms.
Collapse
Affiliation(s)
- Ashraf I Khasawneh
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqa, Jordan
| | - Nisreen Himsawi
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqa, Jordan
| | - Jumana Abu-Raideh
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqa, Jordan
| | - Muna A Salameh
- Department of Basic Medical Sciences, College of Medicine, Al-Balqa' Applied University, Al-Salt, Jordan
| | - Mohammad Al-Tamimi
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqa, Jordan
| | - Sameer Al Haj Mahmoud
- Department of Basic Medical Sciences, College of Medicine, Al-Balqa' Applied University, Al-Salt, Jordan
| | - Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqa, Jordan
| |
Collapse
|
25
|
Lakshmi SA, Bhaskar JP, Krishnan V, Sethupathy S, Pandipriya S, Aruni W, Pandian SK. Inhibition of biofilm and biofilm-associated virulence factor production in methicillin-resistant Staphylococcus aureus by docosanol. J Biotechnol 2020; 317:59-69. [PMID: 32353392 DOI: 10.1016/j.jbiotec.2020.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/25/2020] [Accepted: 04/23/2020] [Indexed: 12/14/2022]
Abstract
Antimicrobial resistance is a major public health concern in infection control. Hence, a multi-pronged approach is necessary to curb the severity of infections. The present study entails the identification of docosanol (fatty alcohol) from Streptomyces as a novel antibiofilm agent which can target the virulence factors of MRSA. Results showed that docosanol as a potent antibiofilm agent and found to inhibit several virulence factors of MRSA. The antibiofilm efficacy of docosanol analyzed through light and scanning electron microscopy showed a significant reduction in adherent cells. Moreover, analysis of three-dimensional structure of biofilm matrix by confocal laser scanning microscope demonstrated effective antibiofilm potential of docosanol. In addition, docosanol reduced the survival rate of MRSA in healthy human blood and enhanced the neutrophil-mediated killing by interfering with hemolysin production. RT-qPCR analysis revealed the down regulation of several virulence genes, possibly by affecting the expression of the accessory gene regulator (agr) system and transcriptional regulator sarA. These findings suggest that docosanol could effectively reduce the biofilm phenotype and virulence production, and thus becomes a promising candidate to treat MRSA infections.
Collapse
Affiliation(s)
- Selvaraj Alagu Lakshmi
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India
| | | | | | - Sivasamy Sethupathy
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India
| | - Selvapandi Pandipriya
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India
| | - Wilson Aruni
- Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India; Musculoskeletal Disease Research Center, US Department of Veteran Affairs, VA, California, USA
| | | |
Collapse
|
26
|
Liu J, Kozhaya L, Torres VJ, Unutmaz D, Lu M. Structure-based discovery of a small-molecule inhibitor of methicillin-resistant Staphylococcus aureus virulence. J Biol Chem 2020; 295:5944-5959. [PMID: 32179646 PMCID: PMC7196633 DOI: 10.1074/jbc.ra120.012697] [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] [Received: 01/16/2020] [Revised: 03/06/2020] [Indexed: 01/07/2023] Open
Abstract
The rapid emergence and dissemination of methicillin-resistant Staphylococcus aureus (MRSA) strains poses a major threat to public health. MRSA possesses an arsenal of secreted host-damaging virulence factors that mediate pathogenicity and blunt immune defenses. Panton-Valentine leukocidin (PVL) and α-toxin are exotoxins that create lytic pores in the host cell membrane. They are recognized as being important for the development of invasive MRSA infections and are thus potential targets for antivirulence therapies. Here, we report the high-resolution X-ray crystal structures of both PVL and α-toxin in their soluble, monomeric, and oligomeric membrane-inserted pore states in complex with n-tetradecylphosphocholine (C14PC). The structures revealed two evolutionarily conserved phosphatidylcholine-binding mechanisms and their roles in modulating host cell attachment, oligomer assembly, and membrane perforation. Moreover, we demonstrate that the soluble C14PC compound protects primary human immune cells in vitro against cytolysis by PVL and α-toxin and hence may serve as the basis for the development of an antivirulence agent for managing MRSA infections.
Collapse
Affiliation(s)
- Jie Liu
- Public Health Research Institute, Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Lina Kozhaya
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032
| | - Victor J. Torres
- Department of Microbiology, New York University School of Medicine, New York, New York 10016
| | - Derya Unutmaz
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032
| | - Min Lu
- Public Health Research Institute, Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, To whom correspondence should be addressed:
Public Health Research Institute, Dept. of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Newark, NJ 07103. Tel.:
973-854-3260; E-mail:
| |
Collapse
|
27
|
Yu S, Jiang B, Jia C, Wu H, Shen J, Hu X, Xie Z. Investigation of biofilm production and its association with genetic and phenotypic characteristics of OM (osteomyelitis) and non-OM orthopedic Staphylococcus aureus. Ann Clin Microbiol Antimicrob 2020; 19:10. [PMID: 32220258 PMCID: PMC7099788 DOI: 10.1186/s12941-020-00352-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 03/13/2020] [Indexed: 11/25/2022] Open
Abstract
Background Staphylococcus aureus is a primary pathogen of orthopedic infections. By mediating antimicrobial resistance, S. aureus biofilm plays an important role in the recalcitrance of orthopedic infections, especially for the intractable osteomyelitis (OM). This study investigated the relationship between biofilm production and various genetic or phenotypic characteristics among orthopedic S. aureus strains. Methods A total of 137 orthopedic S. aureus isolates were enrolled and divided into OM and non-OM groups. Biofilm production was evaluated using the crystal violet assay. Genetic and phenotypic characteristics including MRSA identification, MLST and spa typing, carriage of virulence genes, drug resistance, and patients’ inflammatory responses indicators were characterized. The relationship between biofilm production and above-mentioned features was respectively analyzed among all isolates and compared between OM and non-OM isolates. Results Biofilm production presented no significant difference between OM (including 9 MRSA isolates) and non-OM (including 21 MRSA isolates) strains. We found that ST88, t377 and ST630-MSSA-t377 strains produced very strong biofilms, while MLST types of ST15, ST25, ST398, ST5, ST59 and spa types of t002, t2325, t437 tended to produce weaker biofilms. Strains with the following profiles produced stronger biofilms: fib(+)-hlgv(+)-lukED(+)-sei(-)-sem(-)-seo(-) for all isolates, sei(-)-sem(-)-seo(-) for OM isolates, and cna (+)-fib (+)-hlgv (+)-lukED (+)-seb(-)-sed(-) for non-OM isolates. In addition, not any single drug resistance was found to be related to biofilm production. We also observed that, among OM patients, strains with stronger biofilms caused weaker inflammatory responses. Conclusion Some genetic or phenotypic characteristics of orthopedic strains were associated with biofilm production, and this association could be different among OM and non-OM strains. The results are of great significance for better understanding, evaluating and managing different kinds of biofilm-associated orthopedic infections, and provide potential targets for biofilm clearance.
Collapse
Affiliation(s)
- Shengpeng Yu
- Department of Orthopedics, Southwest Hospital, Army Medical University, Gaotanyan Main Street 30#, District Shapingba, Chongqing, China.,Department of Orthopedics, Dujiangyan Medical Center, Dujiangyan, Sichuan, China
| | - Bei Jiang
- Department of Orthopedics, Southwest Hospital, Army Medical University, Gaotanyan Main Street 30#, District Shapingba, Chongqing, China.,Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Gaotanyan Main Street 30#, District Shapingba, Chongqing, China
| | - Chao Jia
- Department of Orthopedics, Southwest Hospital, Army Medical University, Gaotanyan Main Street 30#, District Shapingba, Chongqing, China
| | - Hongri Wu
- Department of Orthopedics, Southwest Hospital, Army Medical University, Gaotanyan Main Street 30#, District Shapingba, Chongqing, China
| | - Jie Shen
- Department of Orthopedics, Southwest Hospital, Army Medical University, Gaotanyan Main Street 30#, District Shapingba, Chongqing, China
| | - Xiaomei Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Gaotanyan Main Street 30#, District Shapingba, Chongqing, China.
| | - Zhao Xie
- Department of Orthopedics, Southwest Hospital, Army Medical University, Gaotanyan Main Street 30#, District Shapingba, Chongqing, China.
| |
Collapse
|
28
|
Wang C, Chen P, Qiao Y, Kang Y, Yan C, Yu Z, Wang J, He X, Wu H. pH responsive superporogen combined with PDT based on poly Ce6 ionic liquid grafted on SiO 2 for combating MRSA biofilm infection. Theranostics 2020; 10:4795-4808. [PMID: 32308750 PMCID: PMC7163436 DOI: 10.7150/thno.42922] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Biofilm infection caused by multidrug-resistant bacteria is difficult to eradicate by conventional therapies. Photodynamic therapy (PDT) is an effective antibacterial method for fighting against biofilm infection. However, the blocked photosensitizers outside of biofilm greatly limit the efficacy of PDT. Methods: Herein, a novel acid-responsive superporogen and photosensitizer (SiO2-PCe6-IL) was developed. Because of the protonation of the photosensitizer and the high binding energy of the polyionic liquid, SiO2-PCe6-IL changed to positive SiO2-PIL+ in an acidic microenvironment of biofilm infection. SiO2-PIL+ could combine with negatively charged extracellular polymeric substances (EPS) and create holes to remove the biofilm barrier. To strengthen the interaction between SiO2-PIL+ and EPS, SiO2-PIL+ of high charge density was prepared by grafting the high-density initiation site of ATRP onto the surface of the SiO2 base. Results: Due to the rapid protonation rate of COO- and the strong binding energy of SiO2-PIL+ with EPS, SiO2-PCe6-IL could release 90% of Ce6 in 10 s. With the stronger electrostatic and hydrophobic interaction of SiO2-PIL+ with EPS, the surface potential, hydrophobicity, adhesion and mechanical strength of biofilm were changed, and holes in the biofilm were created in 10 min. Combining with the release of photosensitizers and the porous structure of the biofilm, Ce6 was efficiently concentrated in the biofilm. The in vitro and in vivo antibacterial experiments proved that SiO2-PCe6-IL dramatically improved the PDT efficacy against MRSA biofilm infection. Conclusion: These findings suggest that SiO2-PCe6-IL could rapidly increase the concentration of photosensitizer in biofilm and it is an effective therapy for combating biofilm infection.
Collapse
|
29
|
Glycyrrhizin administration ameliorates Streptococcus aureus-induced acute lung injury. Int Immunopharmacol 2019; 70:504-511. [PMID: 30884430 DOI: 10.1016/j.intimp.2019.02.046] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/10/2019] [Accepted: 02/25/2019] [Indexed: 12/25/2022]
Abstract
Streptococcus aureus (S. aureus)-induced acute lung injury (ALI) has a high incidence of mortality clinically. Glycyrrhizin (GL) is a traditional Chinese medicine for anti-inflammatory. However, the role of GL in S. aureus-induced ALI has not previously been elucidated. GL (25 mg/kg i.p.) administration exerts potent anti-inflammatory effect in this model. GL administration significantly alleviated inflammation via reduction of multiple cytokines (serum and lung tissue IL-6, TNF-α, IL-8, IL-1β and HMGB1) and immune cells (lung tissue neutrophil and macrophage infiltration). Additionally, we measured the signaling pathways (NF-kB and MARKs) and inflammasome dependent pyroptosis. The results suggest that GL inhibits NF-kB, p38/ERK pathways and pyroptosis. Furthermore, we used different inhibitors to treat infected-A549 cells and found that BMS-582949 (a p38 inhibitor) is the most effective inhibitor for inhibiting pro-inflammatory cytokines (IL-6, TNF-α and IL-1β) production, which suggests that p38 signaling pathway might be the main pathway for S. aureus-induced inflammation. Collectively, the data demonstrates that GL could mitigate inflammation after S. aureus infection.
Collapse
|
30
|
Raafat D, Otto M, Reppschläger K, Iqbal J, Holtfreter S. Fighting Staphylococcus aureus Biofilms with Monoclonal Antibodies. Trends Microbiol 2019; 27:303-322. [PMID: 30665698 DOI: 10.1016/j.tim.2018.12.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/10/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus (S. aureus) is a notorious pathogen and one of the most frequent causes of biofilm-related infections. The treatment of S. aureus biofilms is hampered by the ability of the biofilm structure to shield bacteria from antibiotics as well as the host's immune system. Therefore, new preventive and/or therapeutic interventions, including the use of antibody-based approaches, are urgently required. In this review, we describe the mechanisms by which anti-S. aureus antibodies can help in combating biofilms, including an up-to-date overview of monoclonal antibodies currently in clinical trials. Moreover, we highlight ongoing efforts in passive vaccination against S. aureus biofilm infections, with special emphasis on promising targets, and finally indicate the direction into which future research could be heading.
Collapse
Affiliation(s)
- Dina Raafat
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Egypt; Current affiliation: Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Kevin Reppschläger
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Jawad Iqbal
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|