1
|
Duan J, Li B, Liu Y, Han T, Ye F, Xia H, Liu K, He J, Wang X, Cai Q, Meng W, Zhu S. Ultra-Photostable Bacterial-Seeking Near-Infrared CPDs for Simultaneous NIR-II Bioimaging and Antibacterial Therapy. Adv Healthc Mater 2024:e2401131. [PMID: 39225395 DOI: 10.1002/adhm.202401131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Bacterial infections can pose significant health risks as they have the potential to cause a range of illnesses. These infections can spread rapidly and lead to complications if not promptly diagnosed and treated. Therefore, it is of great significance to develop a probe to selectively target and image pathogenic bacteria while simultaneously killing them, as there are currently no effective clinical solutions available. This study presents a novel approach using near-infrared carbonized polymer dots (NIR-CPDs) for simultaneous in vivo imaging and treatment of bacterial infections. The core-shell structure of the NIR-CPDs facilitates their incorporation into bacterial cell membranes, leading to an increase in fluorescence brightness and photostability. Significantly, the NIR-CPDs exhibit selective bacterial-targeting properties, specifically identifying Staphylococcus aureus (S. aureus) while sparing Escherichia coli (E. coli). Moreover, under 808 nm laser irradiation, the NIR-CPDs exhibit potent photodynamic effects by generating reactive oxygen species that target and damage bacterial membranes. In vivo experiments on infected mouse models demonstrate not only precise imaging capabilities but also significant therapeutic efficacy, with marked improvements in wound healing. The study provides the dual-functional potential of NIR-CPDs as a highly effective tool for the advancement of medical diagnostics and therapeutics in the fight against bacterial infections.
Collapse
Affiliation(s)
- Jingyi Duan
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Jilin University, Changchun, 130021, P. R. China
| | - Baosheng Li
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Yanqun Liu
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Jilin University, Changchun, 130021, P. R. China
| | - Tianyang Han
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Fengming Ye
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Jilin University, Changchun, 130021, P. R. China
| | - Huan Xia
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Jilin University, Changchun, 130021, P. R. China
| | - Kaifeng Liu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, P. R. China
| | - Jie He
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Jilin University, Changchun, 130021, P. R. China
| | - Xueke Wang
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Jilin University, Changchun, 130021, P. R. China
| | - Qing Cai
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Weiyan Meng
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Shoujun Zhu
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| |
Collapse
|
2
|
Jeong GJ, Khan F, Tabassum N, Kim YM. Natural and synthetic molecules with potential to enhance biofilm formation and virulence properties in Pseudomonas aeruginosa. Crit Rev Microbiol 2024; 50:830-858. [PMID: 37968960 DOI: 10.1080/1040841x.2023.2282459] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 11/17/2023]
Abstract
Pseudomonas aeruginosa can efficiently adapt to changing environmental conditions due to its ubiquitous nature, intrinsic/acquired/adaptive resistance mechanisms, high metabolic versatility, and the production of numerous virulence factors. As a result, P. aeruginosa becomes an opportunistic pathogen, causing chronic infection in the lungs and several organs of patients suffering from cystic fibrosis. Biofilm established by P. aeruginosa in host tissues and medical device surfaces has been identified as a major obstruction to antimicrobial therapy. P. aeruginosa is very likely to be closely associated with the various microorganisms in the host tissues or organs in a pathogenic or nonpathogenic behavior. Aside from host-derived molecules, other beneficial and pathogenic microorganisms produce a diverse range of secondary metabolites that either directly or indirectly favor the persistence of P. aeruginosa. Thus, it is critical to understand how P. aeruginosa interacts with different molecules and ions in the host and abiotic environment to produce extracellular polymeric substances and virulence factors. Thus, the current review discusses how various natural and synthetic molecules in the environment induce biofilm formation and the production of multiple virulence factors.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea
| | - Fazlurrahman Khan
- Institute of Fisheries Sciences, Pukyong National University, Busan, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
3
|
Mirzaei R, Campoccia D, Ravaioli S, Arciola CR. Emerging Issues and Initial Insights into Bacterial Biofilms: From Orthopedic Infection to Metabolomics. Antibiotics (Basel) 2024; 13:184. [PMID: 38391570 PMCID: PMC10885942 DOI: 10.3390/antibiotics13020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
Bacterial biofilms, enigmatic communities of microorganisms enclosed in an extracellular matrix, still represent an open challenge in many clinical contexts, including orthopedics, where biofilm-associated bone and joint infections remain the main cause of implant failure. This study explores the scenario of biofilm infections, with a focus on those related to orthopedic implants, highlighting recently emerged substantial aspects of the pathogenesis and their potential repercussions on the clinic, as well as the progress and gaps that still exist in the diagnostics and management of these infections. The classic mechanisms through which biofilms form and the more recently proposed new ones are depicted. The ways in which bacteria hide, become impenetrable to antibiotics, and evade the immune defenses, creating reservoirs of bacteria difficult to detect and reach, are delineated, such as bacterial dormancy within biofilms, entry into host cells, and penetration into bone canaliculi. New findings on biofilm formation with host components are presented. The article also delves into the emerging and critical concept of immunometabolism, a key function of immune cells that biofilm interferes with. The growing potential of biofilm metabolomics in the diagnosis and therapy of biofilm infections is highlighted, referring to the latest research.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all'Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Stefano Ravaioli
- Laboratorio di Patologia delle Infezioni Associate all'Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Carla Renata Arciola
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| |
Collapse
|
4
|
Ul Haq I, Khan TA, Krukiewicz K. Etiology, pathology, and host-impaired immunity in medical implant-associated infections. J Infect Public Health 2024; 17:189-203. [PMID: 38113816 DOI: 10.1016/j.jiph.2023.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/20/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
Host impaired immunity and pathogens adhesion factors are the key elements in analyzing medical implant-associated infections (MIAI). The infection chances are further influenced by surface properties of implants. This review addresses the medical implant-associated pathogens and summarizes the etiology, pathology, and host-impaired immunity in MIAI. Several bacterial and fungal pathogens have been isolated from MIAI; together, they form cross-kingdom species biofilms and support each other in different ways. The adhesion factors initiate the pathogen's adherence on the implant's surface; however, implant-induced impaired immunity promotes the pathogen's colonization and biofilm formation. Depending on the implant's surface properties, immune cell functions get slow or get exaggerated and cause immunity-induced secondary complications resulting in resistant depression and immuno-incompetent fibro-inflammatory zone that compromise implant's performance. Such consequences lead to the unavoidable and straightforward conclusion for the downstream transformation of new ideas, such as the development of multifunctional implant coatings.
Collapse
Affiliation(s)
- Ihtisham Ul Haq
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland; Joint Doctoral School, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland; Programa de Pós-graduação em Inovação Tecnológica, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil.
| | - Taj Ali Khan
- Division of Infectious Diseases & Global Medicine, Department of Medicine, University of Florida, Gainesville, FL, United States; Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan.
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland; Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego 22B, 44-100 Gliwice, Poland.
| |
Collapse
|
5
|
Hsu CY, Faisal Mutee A, Porras S, Pineda I, Ahmed Mustafa M, J Saadh M, Adil M, H A Z. Amphiregulin in infectious diseases: Role, mechanism, and potential therapeutic targets. Microb Pathog 2024; 186:106463. [PMID: 38036111 DOI: 10.1016/j.micpath.2023.106463] [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: 10/07/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Amphiregulin (AREG) serves as a ligand for the epidermal growth factor receptor (EGFR) and is involved in vital biological functions, including inflammatory responses, tissue regeneration, and immune system function. Upon interaction with the EGFR, AREG initiates a series of signaling cascades necessary for several physiological activities, such as metabolism, cell cycle regulation, and cellular proliferation. Recent findings have provided evidence for the substantial role of AREG in maintaining the equilibrium of homeostasis in damaged tissues and preserving epithelial cell structure in the context of viral infections affecting the lungs. The development of resistance to influenza virus infection depends on the presence of type 1 cytokine responses. Following the eradication of the pathogen, the lungs are subsequently colonized by several cell types that are linked with type 2 immune responses. These cells contribute to the process of repairing and resolving the tissue injury and inflammation caused by infections. Following influenza infection, the activation of AREG promotes the regeneration of bronchial epithelial cells, enhancing the tissue's structural integrity and increasing the survival rate of infected mice. In the same manner, mice afflicted with influenza experience rapid mortality due to a subsequent bacterial infection in the pulmonary region when both bacterial and viral infections manifest concurrently inside the same host. The involvement of AREG in bacterial infections has been demonstrated. The gene AREG experiences increased transcriptional activity inside host cells in response to bacterial infections caused by pathogens such as Escherichia coli and Neisseria gonorrhea. In addition, AREG has been extensively studied as a mitogenic stimulus in epithelial cell layers. Consequently, it is regarded as a prospective contender that might potentially contribute to the observed epithelial cell reactions in helminth infection. Consistent with this finding, mice that lack the AREG gene exhibit a delay in the eradication of the intestinal parasite Trichuris muris. The observed delay is associated with a reduction in the proliferation rate of colonic epithelial cells compared to the infected animals in the control group. The aforementioned findings indicate that AREG plays a pivotal role in facilitating the activation of defensive mechanisms inside the epithelial cells of the intestinal tissue. The precise cellular sources of AREG in this specific context have not yet been determined. However, it is evident that the increased proliferation of the epithelial cell layer in infected mice is reliant on CD4+ T cells. The significance of this finding lies in its demonstration of the crucial role played by the interaction between immunological and epithelial cells in regulating the AREG-EGFR pathway. Additional research is necessary to delve into the cellular origins and signaling mechanisms that govern the synthesis of AREG and its tissue-protective properties, independent of infection.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City 71710, Taiwan
| | | | - Sandra Porras
- Facultad de Mecánica, Escuela Superior Politécnica de Chimborazo (ESPOCH), Panamericana Sur km 1 1/2, Riobamba, 060155, Ecuador
| | - Indira Pineda
- Facultad de Salud Pública, Escuela Superior Politécnica de Chimborazo (ESPOCH), Panamericana Sur km 1 1/2, Riobamba, 060155, Ecuador
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, Imam Jaafar AL-Sadiq University, Iraq; Department of Pathological Analyzes, College of Applied Sciences, University of Samarra, Iraq.
| | - Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan; Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | | | - Zainab H A
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| |
Collapse
|
6
|
El-Tantawy AI, Elmongy EI, Elsaeed SM, Abdel Aleem AAH, Binsuwaidan R, Eisa WH, Salman AU, Elharony NE, Attia NF. Synthesis, Characterization, and Docking Study of Novel Thioureidophosphonate-Incorporated Silver Nanocomposites as Potent Antibacterial Agents. Pharmaceutics 2023; 15:1666. [PMID: 37376114 DOI: 10.3390/pharmaceutics15061666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/27/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Newly synthesized mono- and bis-thioureidophosphonate (MTP and BTP) analogues in eco-friendly conditions were employed as reducing/capping cores for 100, 500, and 1000 mg L-1 of silver nitrate. The physicochemical properties of silver nanocomposites (MTP(BTP)/Ag NCs) were fully elucidated using spectroscopic and microscopic tools. The antibacterial activity of the nanocomposites was screened against six multidrug-resistant pathogenic strains, comparable to ampicillin and ciprofloxacin commercial drugs. The antibacterial performance of BTP was more substantial than MTP, notably with the best minimum inhibitory concentration (MIC) of 0.0781 mg/mL towards Bacillus subtilis, Salmonella typhi, and Pseudomonas aeruginosa. Among all, BTP provided the clearest zone of inhibition (ZOI) of 35 ± 1.00 mm against Salmonella typhi. After the dispersion of silver nanoparticles (AgNPs), MTP/Ag NCs offered dose-dependently distinct advantages over the same nanoparticle with BTP; a more noteworthy decline by 4098 × MIC to 0.1525 × 10-3 mg/mL was recorded for MTP/Ag-1000 against Pseudomonas aeruginosa over BTP/Ag-1000. Towards methicillin-resistant Staphylococcus aureus (MRSA), the as-prepared MTP(BTP)/Ag-1000 displayed superior bactericidal ability in 8 h. Because of the anionic surface of MTP(BTP)/Ag-1000, they could effectively resist MRSA (ATCC-43300) attachment, achieving higher antifouling rates of 42.2 and 34.4% at most optimum dose (5 mg/mL), respectively. The tunable surface work function between MTP and AgNPs promoted the antibiofilm activity of MTP/Ag-1000 by 1.7 fold over BTP/Ag-1000. Lastly, the molecular docking studies affirmed the eminent binding affinity of BTP over MTP-besides the improved binding energy of MTP/Ag NC by 37.8%-towards B. subtilis-2FQT protein. Overall, this study indicates the immense potential of TP/Ag NCs as promising nanoscale antibacterial candidates.
Collapse
Affiliation(s)
- Ahmed I El-Tantawy
- Department of Chemistry, Faculty of Science, Menoufia University, Shibin El Kom 32511, Egypt
| | - Elshaymaa I Elmongy
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Shimaa M Elsaeed
- Department of Analysis and Evaluation, Egyptian Petroleum Research Institute, Cairo 11727, Egypt
| | | | - Reem Binsuwaidan
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Wael H Eisa
- Spectroscopy Department, Physics Division, National Research Centre (NRC), Cairo 12622, Egypt
| | - Ayah Usama Salman
- Department of Botany and Microbiology, Faculty of Science, Menoufia University, Shibin El Kom 32511, Egypt
| | - Noura Elsayed Elharony
- Department of Chemistry, Faculty of Science, Menoufia University, Shibin El Kom 32511, Egypt
| | - Nour F Attia
- Gas Analysis and Fire Safety Laboratory, Chemistry Division, National Institute for Standards, 136, Giza 12211, Egypt
| |
Collapse
|
7
|
Wei Y, Huang N, Ye X, Liu M, Wei M, Huang Y. The postbiotic of hawthorn-probiotic ameliorating constipation caused by loperamide in elderly mice by regulating intestinal microecology. Front Nutr 2023; 10:1103463. [PMID: 37006920 PMCID: PMC10061020 DOI: 10.3389/fnut.2023.1103463] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
Background Constipation is common gastrointestinal disorder with high prevalence and recurrence, making people suffering. However, the treatment for constipation remains ineffectual. We aimed to the study the effects and mechanisms of postbiotic of hawthorn-probiotic on loperamide modeled old KM mice. Methods Constipated mice were grouped and treated with 10% lactulose (Y), hawthorn group (S), probiotic group (F) and postbiotic of hawthorn-probiotic (FS). Fecal changes were observed. AQP3 and Enac-γ were measured by RT-qPCR and Western blotting, intestinal barrier by H&E and immunofluorescence staining, cell proliferation and apoptosis by CCK8 and flow cytometry. Gut microbiota was further determined by 16 s rRNA sequence of feces. Results Postbiotic of hawthorn-probiotic improved intestinal movement and pathomorphology, elevated AQP3, Enac-γ and mucin-2 expression, accompanied by decreased serum TNF-α and cell apoptosis, but increased proliferation. Furthermore, it modified the gut microbiota of constipated mice, featured by upregulation of Lactobacillaceae. Conclusion Postbiotic of hawthorn-probiotic relieved constipation by combined effects of regulating intestinal water and sodium metabolism, maintain intestinal barrier and gut microflora.Graphical Abstract.
Collapse
Affiliation(s)
- Yu Wei
- Basic Medical Science College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Na Huang
- The Eighth School of Clinical Medicine (Foshan Hospital of TCM), Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xinyu Ye
- Basic Medical Science College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Meng Liu
- Basic Medical Science College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Meilian Wei
- Basic Medical Science College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yali Huang
- Basic Medical Science College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Mirzaei R, Esmaeili Gouvarchin Ghaleh H, Ranjbar R. Antibiofilm effect of melittin alone and in combination with conventional antibiotics toward strong biofilm of MDR-MRSA and - Pseudomonas aeruginosa. Front Microbiol 2023; 14:1030401. [PMID: 36910230 PMCID: PMC9994733 DOI: 10.3389/fmicb.2023.1030401] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 02/01/2023] [Indexed: 02/22/2023] Open
Abstract
Introduction Multidrug-resistant (MDR) pathogens are being recognized as a critical threat to human health if they can form biofilm and, in this sense, biofilm-forming MDR-methicillin resistant Staphylococcus aureus (MRSA) and -Pseudomonas aeruginosa strains are a worse concern. Hence, a growing body of documents has introduced antimicrobial peptides (AMPs) as a substitute candidate for conventional antimicrobial agents against drug-resistant and biofilm-associated infections. We evaluated melittin's antibacterial and antibiofilm activity alone and/or in combination with gentamicin, ciprofloxacin, rifampin, and vancomycin on biofilm-forming MDR-P. aeruginosa and MDR-MRSA strains. Methods Antibacterial tests [antibiogram, minimum inhibitory concentration (MIC), and minimum bactericidal concentration (MBC)], anti-biofilm tests [minimum biofilm inhibition concentration (MBIC), and minimum biofilm eradication concentration (MBEC)], as well as synergistic antibiofilm activity of melittin and antibiotics, were performed. Besides, the influence of melittin alone on the biofilm encoding genes and the cytotoxicity and hemolytic effects of melittin were examined. Results MIC, MBC, MBIC, and MBEC indices for melittin were in the range of 0.625-5, 1.25-10, 2.5-20, and 10-40 μg/ml, respectively. The findings found that the combination of melittin AMP with antibiotics was synergistic and fractional biofilm inhibitory concentration index (FBICi) for most tested concentrations was <0.5, resulting in a significant reduction in melittin, gentamicin, ciprofloxacin, vancomycin, and rifampin concentrations by 2-256.4, 2-128, 2-16, 4-64 and 4-8 folds, respectively. This phenomenon reduced the toxicity of melittin, whereby its synergist concentration required for biofilm inhibition did not show cytotoxicity and hemolytic activity. Our findings found that melittin decreased the expression of icaA in S. aureus and LasR in P. aeruginosa genes from 0.1 to 4.11 fold for icaA, and 0.11 to 3.7 fold for LasR, respectively. Conclusion Overall, the results obtained from our study show that melittin alone is effective against the strong biofilm of MDR pathogens and also offers sound synergistic effects with antibiotics without toxicity. Hence, combining melittin and antibiotics can be a potential candidate for further evaluation of in vivo infections by MDR pathogens.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Reza Ranjbar
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
The Emerging Role of Probiotics and their Derivatives against Biofilm-Producing MRSA: A Scoping Review. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4959487. [PMID: 36605101 PMCID: PMC9810406 DOI: 10.1155/2022/4959487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/20/2022] [Accepted: 12/01/2022] [Indexed: 12/29/2022]
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) is one of the main bacterial pathogens causing chronic infections, mainly because of its capacity to produce biofilm. Biofilm production is one of the underlying strategies for antibacterial drug resistance. Accordingly, preventing and attenuating biofilm production has become an emerging approach to controlling persistent infections. Therefore, this scoping review is aimed at surveying the published literature describing the usage of probiotics and their derivatives against biofilm-producing MRSA. Methods Updated literature searches were conducted across seven electronic databases including Web of Science, PubMed, Scopus, Cochrane Library, ProQuest, Embase, and Google Scholar to identify all original published articles about probiotics against MRSA. In this regard, studies were summarized and analyzed in the present review. Results In the reviewed studies, various microorganisms and compounds were used as probiotics as follows: Lactobacillus species (8 studies), Enterococcus species (4 studies), Bacillus species (2 studies), Streptomyces species (2 studies), Saccharomyces cerevisiae (1 study), Corynebacterium accolens (1 study), and Lactococcus lactis derived Nisin (3 studies). Based on our comprehensive search, 21 studies with eligibility criteria were included in the present review including 12 studies on clinical strains, 6 studies on ATCC, 2 studies simultaneously on clinical and standard strains, and finally 1 study on food sample. Conclusions Our study showed that there was an increasing trend in the number of publications reporting probiotics against biofilm-producing MRSA. The results of this scoping review could use to guide the undertaking of the subsequent systematic reviews. In summary, probiotics with antimicrobial and antibiofilm properties can use as an embedded agent in food products or as a biopharmaceutical in the prevention and treatment of MRSA infections.
Collapse
|
10
|
Mirzaei R, Yousefimashouf R, Arabestani MR, Sedighi I, Alikhani MY. The issue beyond resistance: Methicillin-resistant Staphylococcus epidermidis biofilm formation is induced by subinhibitory concentrations of cloxacillin, cefazolin, and clindamycin. PLoS One 2022; 17:e0277287. [PMID: 36350834 PMCID: PMC9645612 DOI: 10.1371/journal.pone.0277287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
Staphylococcus epidermis is one of the most frequent causes of device-associated infections due to biofilm formation. Current reports noted that subinhibitory concentrations of antibiotics induce biofilm production in some bacteria. Accordingly, we evaluated the effect of exposure of different subinhibitory concentrations of cloxacillin, cefazolin, clindamycin, and vancomycin on the biofilm formation of methicillin-resistant S. epidermidis (MRSE). Antimicrobial susceptibility testing and minimum inhibitory/bactericidal concentration of antimicrobial agents were determined. MRSE isolates were selected, and their biofilm formation ability was evaluated. The effect of subinhibitory concentrations of cloxacillin, cefazolin, clindamycin, and vancomycin, antibiotics selected among common choices in the clinic, on MRSE biofilm formation was determined by the microtitre method. Besides, the effect of subinhibitory concentrations of cloxacillin, cefazolin, clindamycin, and vancomycin on the expression of the biofilm-associated genes icaA and atlE was evaluated by Reverse-transcription quantitative real-time polymerase chain reaction (RT-qPCR). Antimicrobial susceptibility patterns of MRSE strains showed a high level of resistance as follows: 80%, 53.3%, 33.3%, 33.3%, and 26.6%, for erythromycin, trimethoprim-sulfamethoxazole, tetracycline, clindamycin, and gentamicin, respectively. Besides, 73.3% of S. epidermidis strains were Multidrug-resistant (MDR). Minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values were in the range of 0.5 to512 μg/mL and 1 to1024 μg/mL for cloxacillin, 0.125 to256 μg/mL and 1 to512 μg/mL for cefazolin, 0.125 to64 μg/mL and 4 to>1024 μg/mL for clindamycin, and 2 to32 μg/mL and 4 to32 μg/mL for vancomycin, respectively. The findings showed that subinhibitory concentrations of cloxacillin, cefazolin, and clindamycin induce biofilm production in MRSE strains. In particular, the OD values of strains were in the range of 0.09-0.95, 0.05-0.86, and 0.06-1 toward cloxacillin, cefazolin, and clindamycin, respectively. On the other hand, exposure to subinhibitory vancomycin concentrations did not increase the biofilm formation in MRSE strains. The findings also demonstrated that sub-MIC of antibiotics up-regulated biofilm-associated genes. In particular, atlE and icaA were up-regulated 0.062 to 1.16 and 0.078 to 1.48 folds, respectively, for cloxacillin, 0.11 to 0.8, and 0.1 to 1.3 folds for cefazolin, 0.18 to 0.98, and 0.19 to 1.4 folds, respectively, for clindamycin. In contrast, the results showed that sub-MIC of vancomycin did not increase the biofilm-associated genes. These findings overall show that exposure to sub-MIC of traditional antibiotics can cause biofilm induction in MRSE, thereby increasing the survival and persistence on various surfaces that worsen the condition of comorbid infections.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Reza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Sedighi
- Department of Pediatrics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Brucellosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
11
|
Negm NA, Altalhi AA, Saleh Mohamed NE, Kana MTHA, Mohamed EA. Growth Inhibition of Sulfate-Reducing Bacteria during Gas and Oil Production Using Novel Schiff Base Diquaternary Biocides: Synthesis, Antimicrobial, and Toxicological Assessment. ACS OMEGA 2022; 7:40098-40108. [PMID: 36385895 PMCID: PMC9647739 DOI: 10.1021/acsomega.2c04836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
Upstream crude oil production equipment is always exposed to destruction damagingly which is caused by sulfate-reducing bacterium (SRB) activities that produce H2S gas, which leads to increased metal corrosion (bio-fouling) rates and inflicts effective infrastructure damage. Hence, oil and gas reservoirs must be injected with biocides and inhibitors which still offer the foremost protection against harmful microbial activity. However, because of the economic and environmental risks associated with biocides, the oil and gas sectors improve better methods for their usage. This work describes the synthesis and evaluation of the biological activities as the cytotoxicity and antimicrobial properties of a series of diquaternary cationic biocides that were studied during the inhibition of microbial biofilms. The prepared diquaternary compound was synthesized by coupling vanillin and 4-aminoantipyrene to achieve the corresponding Schiff base, followed by a quaternization reaction using 1,6-bromohexane, 1,8-bromooctane, and 1,12-bromododecane. The increase of their alkyl chain length from 6 to 12 methylene groups increased the obtained antimicrobial activity and cytotoxicity. Antimicrobial efficacies of Q1-3 against various biofilm-forming microorganisms, including bacteria and fungi, were examined utilizing the diameter of inhibition zone procedures. The results revealed that cytotoxic efficacies of Q1-3 were significantly associated mainly with maximum surface excess and interfacial characteristics. The cytotoxic efficiencies of Q1-3 biocides demonstrated promising results due to their comparatively higher efficacies against SRB. Q3 exhibited the highest cytotoxic biocide against the gram +ve, gram -ve, and SRB species according to the inhibition zone diameter test. The toxicity of the studied microorganisms depended on the nature and type of the target microorganism and the hydrophobicity of the biocide molecules. Cytotoxicity assessment and antimicrobial activity displayed increased activity by the increase in their alkyl chain length.
Collapse
Affiliation(s)
- Nabel A. Negm
- Egyptian
Petroleum Research Institute, Petrochemicals, 1 Ahmed Elzommer Street, Nasr City, CairoEG 11776, Egypt
| | - Amal A. Altalhi
- Department
of Chemistry, College of Science, Taif University, P.O. Box 11099, Taif21944, Saudi Arabia
| | - Nermin E. Saleh Mohamed
- Egyptian
Petroleum Research Institute, Petrochemicals, 1 Ahmed Elzommer Street, Nasr City, CairoEG 11776, Egypt
| | - Maram T. H. A. Kana
- National
Institute of LASER Enhanced Science, Cairo
University, Giza11776, Egypt
| | - Eslam A. Mohamed
- Egyptian
Petroleum Research Institute, Petrochemicals, 1 Ahmed Elzommer Street, Nasr City, CairoEG 11776, Egypt
| |
Collapse
|
12
|
Wang W, Bao X, Bové M, Rigole P, Meng X, Su J, Coenye T. Antibiofilm Activities of Borneol-Citral-Loaded Pickering Emulsions against Pseudomonas aeruginosa and Staphylococcus aureus in Physiologically Relevant Chronic Infection Models. Microbiol Spectr 2022; 10:e0169622. [PMID: 36194139 PMCID: PMC9602683 DOI: 10.1128/spectrum.01696-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/08/2022] [Indexed: 12/31/2022] Open
Abstract
Phytochemicals are promising antibacterials for the development of novel antibiofilm drugs, but their antibiofilm activity in physiologically relevant model systems is poorly characterized. As the host microenvironment can interfere with the activity of the phytochemicals, mimicking the complex environment found in biofilm associated infections is essential to predict the clinical potential of novel phytochemical-based antimicrobials. In the present study, we examined the antibiofilm activity of borneol, citral, and combinations of both as well as their Pickering emulsions against Staphylococcus aureus and Pseudomonas aeruginosa in an in vivo-like synthetic cystic fibrosis medium (SCFM2) model, an in vitro wound model (consisting of an artificial dermis and blood components at physiological levels), and an in vivo Galleria mellonella model. The Pickering emulsions demonstrated an enhanced biofilm inhibitory activity compared to both citral and the borneol/citral combination, reducing the minimum biofilm inhibitory concentration (MBIC) values up to 2 to 4 times against P. aeruginosa PAO1 and 2 to 8 times against S. aureus P8-AE1 in SCMF2. In addition, citral, the combination borneol/citral, and their Pickering emulsions can completely eliminate the established biofilm of S. aureus P8-AE1. The effectiveness of Pickering emulsions was also demonstrated in the wound model with a reduction of up to 4.8 log units in biofilm formation by S. aureus Mu50. Furthermore, citral and Pickering emulsions exhibited a significant degree of protection against S. aureus infection in the G. mellonella model. The present findings reveal the potential of citral- or borneol/citral-based Pickering emulsions as a type of alternative antibiofilm candidate to control pathogenicity in chronic infection. IMPORTANCE There is clearly an urgent need for novel formulations with antimicrobial and antibiofilm activity, but while there are plenty of studies investigating them using simple in vitro systems, there is a lack of studies in which (combinations of) phytochemicals are evaluated in relevant models that closely resemble the in vivo situation. Here, we examined the antibiofilm activity of borneol, citral, and their combination as well as Pickering emulsions (stabilized by solid particles) of these compounds. Activity was tested against Staphylococcus aureus and Pseudomonas aeruginosa in in vitro models mimicking cystic fibrosis sputum and wounds as well as in an in vivo Galleria mellonella model. The Pickering emulsions showed drastically increased antibiofilm activity compared to that of the compounds as such in both in vitro models and protected G. mellonella larvae from S. aureus-induced killing. Our data show that Pickering emulsions from phytochemicals are potentially useful for treating specific biofilm-related chronic infections.
Collapse
Affiliation(s)
- Wen Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
- China-Singapore International Joint Research Institute, Guangzhou, China
| | - Xuerui Bao
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Mona Bové
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Petra Rigole
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Xiaofeng Meng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- China-Singapore International Joint Research Institute, Guangzhou, China
| | - Jianyu Su
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- China-Singapore International Joint Research Institute, Guangzhou, China
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
13
|
Egorova DA, Solovyev AI, Polyakov NB, Danilova KV, Scherbakova AA, Kravtsov IN, Dmitrieva MA, Rykova VS, Tutykhina IL, Romanova YM, Gintsburg AL. Biofilm matrix proteome of clinical strain of P. aeruginosa isolated from bronchoalveolar lavage of patient in intensive care unit. Microb Pathog 2022; 170:105714. [PMID: 35973647 DOI: 10.1016/j.micpath.2022.105714] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/30/2022] [Accepted: 08/04/2022] [Indexed: 10/15/2022]
Abstract
Extracellular matrix plays a pivotal role in biofilm biology and proposed as a potential target for therapeutics development. As matrix is responsible for some extracellular functions and influence bacterial cytotoxicity against eukaryotic cells, it must have unique protein composition. P. aeruginosa is one of the most important pathogens with emerging antibiotic resistance, but only a few studies were devoted to matrix proteomes and there are no studies describing matrix proteome for any clinical isolates except reference strains PAO1 and ATCC27853. Here we report the first biofilm matrix proteome of P. aeruginosa isolated from bronchoalveolar lavage of patient in intensive care unit. We have identified the largest number of proteins in the matrix among all published studies devoted to P. aeruginosa biofilms. Comparison of matrix proteome with proteome from embedded cells let us to identify several enriched bioprocess groups. Bioprocess groups with the largest number of overrepresented in matrix proteins were oxidation-reduction processes, proteolysis, and transmembrane transport. The top three represented in matrix bioprocesses concerning the size of the GO annotated database were cell redox homeostasis, nucleoside metabolism, and fatty acid synthesis. Finally, we discuss the obtained data in a prism of antibiofilm therapeutics development.
Collapse
Affiliation(s)
- Daria A Egorova
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1).
| | - Andrey I Solovyev
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1)
| | - Nikita B Polyakov
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1)
| | - Ksenya V Danilova
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1)
| | - Anastasya A Scherbakova
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1)
| | - Ivan N Kravtsov
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1)
| | - Maria A Dmitrieva
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1)
| | - Valentina S Rykova
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1)
| | - Irina L Tutykhina
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1)
| | - Yulia M Romanova
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1); I.M. Sechenov First Moscow State Medical University, Moscow, 119992, Russia(2)
| | - Alexander L Gintsburg
- National Research Center of Epidemiology and Microbiology n. a. N.F. Gamaleya, Russian Ministry of Health, Moscow, 123098, Russia(1); I.M. Sechenov First Moscow State Medical University, Moscow, 119992, Russia(2)
| |
Collapse
|
14
|
Rudiansyah M, Jasim SA, Mohammad Pour ZG, Athar SS, Jeda AS, Doewes RI, Jalil AT, Bokov DO, Mustafa YF, Noroozbeygi M, Karampoor S, Mirzaei R. Coronavirus disease 2019 (COVID-19) update: From metabolic reprogramming to immunometabolism. J Med Virol 2022; 94:4611-4627. [PMID: 35689351 PMCID: PMC9350347 DOI: 10.1002/jmv.27929] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/22/2022] [Accepted: 06/09/2022] [Indexed: 12/15/2022]
Abstract
The field of immunometabolism investigates and describes the effects of metabolic rewiring in immune cells throughout activation and the fates of these cells. Recently, it has been appreciated that immunometabolism plays an essential role in the progression of viral infections, cancer, and autoimmune diseases. Regarding COVID‐19, the aberrant immune response underlying the progression of diseases establishes two major respiratory pathologies, including acute respiratory distress syndrome (ARDS) or pneumonia‐induced acute lung injury (ALI). Both innate and adaptive immunity (T cell‐based) were impaired in the course of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infection. Current findings have deciphered that macrophages (innate immune cells) are involved in the inflammatory response seen in COVID‐19. It has been demonstrated that immune system cells can change metabolic reprogramming in some conditions, including autoimmune diseases, cancer, and infectious disease, including COVID‐19. The growing findings on metabolic reprogramming in COVID‐19 allow an exploration of metabolites with immunomodulatory properties as future therapies to combat this hyperinflammatory response. The elucidation of the exact role and mechanism underlying this metabolic reprograming in immune cells could help apply more precise approaches to initial diagnosis, prognosis, and in‐hospital therapy. This report discusses the latest findings from COVID‐19 on host metabolic reprogramming and immunometabolic responses.
Collapse
Affiliation(s)
- Mohammad Rudiansyah
- Division of Nephrology & Hypertension, Department of Internal Medicine, Faculty of Medicine, Universitas Lambung Mangkurat/Ulin Hospital, Banjarmasin, Indonesia
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-Maarif University College, Al-Anbar-Ramadi, Iraq
| | | | - Sara Sohrabi Athar
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran.,Department of Human Nutrition, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Salimi Jeda
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rumi Iqbal Doewes
- Faculty of Sport, Universitas Sebelas Maret, Kentingan, Surakarta, Indonesia
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, Iraq
| | - D O Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation.,Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russian Federation
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Mina Noroozbeygi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
15
|
Guerra MES, Destro G, Vieira B, Lima AS, Ferraz LFC, Hakansson AP, Darrieux M, Converso TR. Klebsiella pneumoniae Biofilms and Their Role in Disease Pathogenesis. Front Cell Infect Microbiol 2022; 12:877995. [PMID: 35646720 PMCID: PMC9132050 DOI: 10.3389/fcimb.2022.877995] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/13/2022] [Indexed: 12/17/2022] Open
Abstract
The ability to form biofilms is a crucial virulence trait for several microorganisms, including Klebsiella pneumoniae – a Gram-negative encapsulated bacterium often associated with nosocomial infections. It is estimated that 65-80% of bacterial infections are biofilm related. Biofilms are complex bacterial communities composed of one or more species encased in an extracellular matrix made of proteins, carbohydrates and genetic material derived from the bacteria themselves as well as from the host. Bacteria in the biofilm are shielded from immune responses and antibiotics. The present review discusses the characteristics of K. pneumoniae biofilms, factors affecting biofilm development, and their contribution to infections. We also explore different model systems designed to study biofilm formation in this species. A great number of factors contribute to biofilm establishment and maintenance in K. pneumoniae, which highlights the importance of this mechanism for the bacterial fitness. Some of these molecules could be used in future vaccines against this bacterium. However, there is still a lack of in vivo models to evaluate the contribution of biofilm development to disease pathogenesis. With that in mind, the combination of different methodologies has great potential to provide a more detailed scenario that more accurately reflects the steps and progression of natural infection.
Collapse
Affiliation(s)
- Maria Eduarda Souza Guerra
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Giulia Destro
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Brenda Vieira
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Alice S. Lima
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Lucio Fabio Caldas Ferraz
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Anders P. Hakansson
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University, Malmo, Sweden
| | - Michelle Darrieux
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Thiago Rojas Converso
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
- *Correspondence: Thiago Rojas Converso,
| |
Collapse
|
16
|
Polymeric Coatings and Antimicrobial Peptides as Efficient Systems for Treating Implantable Medical Devices Associated-Infections. Polymers (Basel) 2022; 14:polym14081611. [PMID: 35458361 PMCID: PMC9024559 DOI: 10.3390/polym14081611] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 02/04/2023] Open
Abstract
Many infections are associated with the use of implantable medical devices. The excessive utilization of antibiotic treatment has resulted in the development of antimicrobial resistance. Consequently, scientists have recently focused on conceiving new ways for treating infections with a longer duration of action and minimum environmental toxicity. One approach in infection control is based on the development of antimicrobial coatings based on polymers and antimicrobial peptides, also termed as “natural antibiotics”.
Collapse
|
17
|
Mirzaei R, Sabokroo N, Ahmadyousefi Y, Motamedi H, Karampoor S. Immunometabolism in biofilm infection: lessons from cancer. Mol Med 2022; 28:10. [PMID: 35093033 PMCID: PMC8800364 DOI: 10.1186/s10020-022-00435-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Biofilm is a community of bacteria embedded in an extracellular matrix, which can colonize different human cells and tissues and subvert the host immune reactions by preventing immune detection and polarizing the immune reactions towards an anti-inflammatory state, promoting the persistence of biofilm-embedded bacteria in the host. MAIN BODY OF THE MANUSCRIPT It is now well established that the function of immune cells is ultimately mediated by cellular metabolism. The immune cells are stimulated to regulate their immune functions upon sensing danger signals. Recent studies have determined that immune cells often display distinct metabolic alterations that impair their immune responses when triggered. Such metabolic reprogramming and its physiological implications are well established in cancer situations. In bacterial infections, immuno-metabolic evaluations have primarily focused on macrophages and neutrophils in the planktonic growth mode. CONCLUSION Based on differences in inflammatory reactions of macrophages and neutrophils in planktonic- versus biofilm-associated bacterial infections, studies must also consider the metabolic functions of immune cells against biofilm infections. The profound characterization of the metabolic and immune cell reactions could offer exciting novel targets for antibiofilm therapy.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Niloofar Sabokroo
- Department of Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Motamedi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|