1
|
Shah I, Uddin Z, Hussain M, Khalil AAK, Amin A, Hanif F, Ali L, Amirzada MI, Shah TA, Dawoud TM, Bourhia M, Li WJ, Sajjad W. Streptomyces sp. from desert soil as a biofactory for antioxidants with radical scavenging and iron chelating potential. BMC Microbiol 2024; 24:419. [PMID: 39434054 PMCID: PMC11492556 DOI: 10.1186/s12866-024-03586-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024] Open
Abstract
Iron homeostasis is vital for normal physiology, but in the majority of circumstances, like iron overload, this equilibrium is upset leading to free iron in the plasma. This condition with excess iron is known as hemochromatosis, which has been linked to many side effects, including cancer and liver cirrhosis. The current research aimed to investigate active molecules from Streptomyces sp. isolated from the extreme environment of Bahawalpur deserts. The strain was characterized using 16 S rRNA sequencing. Chemical analysis of the ethyl acetate cure extract revealed the presence of phenols, flavonoids, alkaloids, and tannins. Multiple ultraviolet (UV) active metabolites that were essential for the stated pharmacological activities were also demonstrated by thin layer chromatography (TLC) and high-performance liquid chromatography (HPLC). Additionally, Gas chromatography/mass spectrometry (GC-MS) analysis revealed the primary constituents of the extract to compose of phenol and ester compounds. The 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay was used to assess the extract's antioxidant capacity, and the results showed a good half-maximal inhibitory concentration (IC50) value of 0.034 µg/mL in comparison to the positive control ascorbic acid's 0.12 µg/mL. In addition, iron chelation activity of extract showed significant chelation potential at 250 and 125 µg/mL, while 62.5 µg/mL showed only mild chelation of the ferrous ion using ethylene diamine tetra acetic acid (EDTA) as a positive control. Likewise, the extract's cytotoxicity was analyzed through 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay using varying concentrations of the extract and showed 51% cytotoxicity at 350 µg/mL and 65% inhibition of cell growth at 700 µg/mL, respectively. The bioactive compounds from Streptomyces sp. demonstrated strong antioxidant and iron chelating potentials and can prolong the cell survival in extreme environment.
Collapse
Affiliation(s)
- Imran Shah
- Department of Pharmacy, Comsats University Islamabad, Abbottabad Campus, Islamabad, Pakistan
| | - Zia Uddin
- Department of Pharmacy, Comsats University Islamabad, Abbottabad Campus, Islamabad, Pakistan
| | - Maheer Hussain
- Department of Biological Sciences, National University of Medical Sciences, Punjab, 46000, Pakistan
| | - Atif Ali Khan Khalil
- Department of Pharmacognosy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Lahore, Pakistan
| | - Arshia Amin
- Department of Biosciences, Capital University of Science and Technology Islamabad, Islamabad, Pakistan
| | - Faisal Hanif
- Army Medical College, National University of Medical Sciences, Rawalpindi, 46000, Pakistan
| | - Liaqat Ali
- Department of Biological Sciences, National University of Medical Sciences, Punjab, 46000, Pakistan
| | - Muhammad Imran Amirzada
- Department of Pharmacy, Comsats University Islamabad, Abbottabad Campus, Islamabad, Pakistan
| | - Tawaf Ali Shah
- College of Agriculture Engineering and Food Science, Shandong University of Technology, Zibo, 255000, China
| | - Turki M Dawoud
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. BOX 2455, Riyadh, 11451, Saudi Arabia
| | - Mohammed Bourhia
- Department of Chemistry and Biochemistry, Faculty of Medicine and Pharmacy, IbnZohr University, Laayoune, 70000, Morocco
| | - Wen-Jun Li
- State Key Laboratory of Desert and Oasis Ecology, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi, 830011, China
- School of Life Sciences, Sun Yat-Sen University, Xingang West Road, Guangzhou, 510275, China
| | - Wasim Sajjad
- Department of Biological Sciences, National University of Medical Sciences, Punjab, 46000, Pakistan.
- State Key Laboratory of Desert and Oasis Ecology, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi, 830011, China.
| |
Collapse
|
2
|
O'Ferrall LM, Fantasia A, Chan K, Teixeira LM, Kavanagh K, O'Connor C, Santos MA, Chaves S, Nurchi VM, Crisponi G, Zoroddu MA, Griffith DM, Cappai R. Solution studies, synthesis and antibacterial activity of Ga(III) complexes with bis-kojate derivatives. J Inorg Biochem 2024; 259:112663. [PMID: 39024775 DOI: 10.1016/j.jinorgbio.2024.112663] [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: 05/31/2024] [Revised: 07/03/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Given the recognized major problem of microbial drug resistance for human health, new metal-based drugs have been currently explored for their antimicrobial properties, including gallium-based compounds as potential metallophores that could perturb Fe's interactions with proteins. Herein we have designed and synthesized two bis-kojate ligands (named L4 and L6) and studied their Ga(III) complexes for their physico-chemical and biological properties. In particular a detailed study of their complexation properties in aqueous solution, showed equilibrium models with formation of quite stable dinuclear 2:3 metal:ligand complexes, though with different stability. Solid state complexes were also prepared and characterized and complementary DFT studies indicated that [Ga2(L4)3] complex, with higher stability, seems to adopt a three-ligand bridging conformation, while that for L6 adopt a one ligand bridging conformation. Preliminary investigation of the antibacterial activity of these gallium complexes showed antipseudomonal activity, which appeared higher for the complex with L4, a feature of potential interest for the scientific community.
Collapse
Affiliation(s)
- L More O'Ferrall
- Department of Chemistry, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland; School of Food Science & Environmental Health, Technological University of Dublin, Dublin 7, Ireland
| | - A Fantasia
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, via Vienna 2, 07100 Sassari, Italy; Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato-Cagliari, Italy
| | - K Chan
- Department of Chemistry, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - L M Teixeira
- Department of Chemistry, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - K Kavanagh
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland; SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland
| | - C O'Connor
- School of Food Science & Environmental Health, Technological University of Dublin, Dublin 7, Ireland
| | - M A Santos
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Engenharia Química, Insituto Superior Técnico, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal
| | - S Chaves
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Engenharia Química, Insituto Superior Técnico, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal
| | - V M Nurchi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato-Cagliari, Italy
| | - G Crisponi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato-Cagliari, Italy
| | - M A Zoroddu
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, via Vienna 2, 07100 Sassari, Italy
| | - D M Griffith
- Department of Chemistry, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland; SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland.
| | - R Cappai
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, via Vienna 2, 07100 Sassari, Italy.
| |
Collapse
|
3
|
Kircheva N, Dobrev S, Nikolova V, Yocheva L, Angelova S, Dudev T. Implementation of Three Gallium-Based Complexes in the "Trojan Horse" Antibacterial Strategy against A. baumannii: A DFT Approach. Inorg Chem 2024; 63:15409-15420. [PMID: 39116415 DOI: 10.1021/acs.inorgchem.4c02411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Microorganisms of the ESKAPE group pose an enormous threat to human well-being, thus requiring a multidisciplinary approach for discovering novel drugs that are not only effective but utilize an innovative mechanism of action in order to decrease fast developing resistance. A promising but still hardly explored implementation in the "Trojan horse" antibacterial strategy has been recognized in gallium, an iron mimicry species with no known function but exerting a bacteriostatic/bactericidal effect against some representatives of the group. The study herewith focuses on the bacterium A. baumannii and its siderophore acinetobactin in its two isomeric forms depending on the acidity of the medium. By applying the powerful tools of the DFT approach, we aim to delineate those physicochemical characteristics that are of great importance for potentiating gallium's ability to compete with the native ferric cation for binding acinetobactin such as pH, solvent exposure (dielectric constant of the environment), different metal/siderophore ratios, and complex composition. Hence, the provided results not only furnish some explanation of the positive effect of three Ga3+-based anti-infectives in terms of metal cation competition but also shed light on reported in vitro and in vivo observations at a molecular level in regard to gallium's antibacterial effect against A. baumannii.
Collapse
Affiliation(s)
- Nikoleta Kircheva
- Institute of Optical Materials and Technologies "Acad. J. Malinowski", Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Stefan Dobrev
- Institute of Optical Materials and Technologies "Acad. J. Malinowski", Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Valya Nikolova
- Faculty of Chemistry and Pharmacy, Sofia University "St. Kl. Ohridski", 1164 Sofia, Bulgaria
| | - Lyubima Yocheva
- Faculty of Chemistry and Pharmacy, Sofia University "St. Kl. Ohridski", 1164 Sofia, Bulgaria
| | - Silvia Angelova
- Institute of Optical Materials and Technologies "Acad. J. Malinowski", Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
- University of Chemical Technology and Metallurgy, 8 St. Kliment Ohridski Blvd, 1756 Sofia, Bulgaria
| | - Todor Dudev
- Faculty of Chemistry and Pharmacy, Sofia University "St. Kl. Ohridski", 1164 Sofia, Bulgaria
| |
Collapse
|
4
|
Meyers M, Salmon M, Libert I, Klášterský J. A meta-analysis on the risk of infection associated with intravenous iron therapy in cancer-associated anaemia: a double-edged sword? Curr Opin Oncol 2024; 36:223-232. [PMID: 38842015 DOI: 10.1097/cco.0000000000001024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
PURPOSE OF REVIEW The increased use of i.v. iron in the treatment of cancer-associated anemia raises concerns about its risk of infectious complications. High levels of circulating iron could increase the risk of infection by compromising natural defence mechanisms and promoting pathogen growth. Since the risk of infection is particularly high in the oncological population, we have examined whether the use of i.v. iron increases the risk of infectious complications among cancer patients. FINDINGS Among 18 randomized trials in our systematic review, only 8 reported infectious complications, with no significant difference linked to the type of i.v. iron preparation. Two trials showed a statistically significant increase in infectious complications, one trial found a lower risk, while the remaining 5 reported no significant difference. Our meta-analysis revealed a numerical increase in infectious complications in the i.v. iron group, but the lack of statistical significance and significant heterogeneity among the trials limit definitive conclusions on the actual infection risk. SUMMARY Our findings suggest some increased risk in infectious complications after the administration of i.v. iron for cancer associated anaemia. However, i.v. iron therapy appears generally safe and effective in cancer-associated anaemia.
Collapse
Affiliation(s)
| | - Maurine Salmon
- Data Centre, Institut Jules Bordet and Université Libre de Bruxelles (ULB)
| | - Isabelle Libert
- Medical Oncology, Supportive Care Unit, Institut Jules Bordet, Brussels, Belgium
| | - Jean Klášterský
- Medical Oncology, Supportive Care Unit, Institut Jules Bordet, Brussels, Belgium
| |
Collapse
|
5
|
Zhang X, Nickerson R, Burton L, Stueck A, Holbein B, Cheng Z, Zhou J, Lehmann C. The Hydroxypyridinone Iron Chelator DIBI Reduces Bacterial Load and Inflammation in Experimental Lung Infection. Biomedicines 2024; 12:1452. [PMID: 39062025 PMCID: PMC11274704 DOI: 10.3390/biomedicines12071452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Iron plays a critical role in lung infections due to its function in the inflammatory immune response but also as an important factor for bacterial growth. Iron chelation represents a potential therapeutic approach to inhibit bacterial growth and pathologically increased pro-inflammatory mediator production. The present study was designed to investigate the impact of the iron chelator DIBI in murine lung infection induced by intratracheal Pseudomonas aeruginosa (strain PA14) administration. DIBI is a polymer with a polyvinylpyrrolidone backbone containing nine 3-hydroxy-1-(methacrylamidoethyl)-2-methyl-4(1H) pyridinone (MAHMP) residues per molecule and was given by intraperitoneal injection either as a single dose (80 mg/kg) immediately after PA14 administration or a double dose (second dose 4 h after PA14 administration). The results showed that lung NF-κBp65 levels, as well as levels of various inflammatory cytokines (TNFα, IL-1β, IL-6) both in lung tissue and bronchoalveolar lavage fluid (BALF), were significantly increased 24 h after PA14 administration. Single-dose DIBI did not affect the bacterial load or inflammatory response in the lungs or BALF. However, two doses of DIBI significantly decreased bacterial load, attenuated NF-κBp65 upregulation, reduced inflammatory cytokines production, and relieved lung tissue damage. Our findings support the conclusion that the iron chelator, DIBI, can reduce lung injury induced by P. aeruginosa, via its anti-bacterial and anti-inflammatory effects.
Collapse
Affiliation(s)
- Xiyang Zhang
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 1X5, Canada; (X.Z.); (J.Z.)
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Precision Anaesthesia and Perioperative Organ Protection, Guangzhou 510515, China
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (R.N.); (L.B.); (B.H.); (Z.C.)
| | - Rhea Nickerson
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (R.N.); (L.B.); (B.H.); (Z.C.)
| | - Lauren Burton
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (R.N.); (L.B.); (B.H.); (Z.C.)
| | - Ashley Stueck
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1X5, Canada;
| | - Bruce Holbein
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (R.N.); (L.B.); (B.H.); (Z.C.)
| | - Zhenyu Cheng
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (R.N.); (L.B.); (B.H.); (Z.C.)
| | - Juan Zhou
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 1X5, Canada; (X.Z.); (J.Z.)
| | - Christian Lehmann
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 1X5, Canada; (X.Z.); (J.Z.)
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (R.N.); (L.B.); (B.H.); (Z.C.)
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 1X5, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
6
|
Song G, Li M, Zhou B, Qi H, Guo J. Gallium-based metal-organic frameworks with antibacterial and anti-inflammatory properties for oral health protection. Heliyon 2024; 10:e31788. [PMID: 38845911 PMCID: PMC11153188 DOI: 10.1016/j.heliyon.2024.e31788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
The fascial space of the oral and maxillofacial region contains loose connective tissues, which possess weak anti-infection ability and are often prone to infection, leading to acute suppurative inflammation and sepsis through blood. Although antibiotic use can reduce the probability of bacterial infections, owing to the emergence of antibiotic-resistant bacteria, the search for new antimicrobial drugs is imminent. Herein, we report a metal-organic framework (MOF) antibacterial material designed and synthesized with gallium (Ga) as the central atom, which possesses significant antibacterial, anti-inflammatory, and antioxidant effects. Our data suggested that GA-based MOFs (Ga-MOFs; 1 μg/mL) could sufficiently kill Porphyromonas gingivalis, Streptococcus pyogenes, and Staphylococcus aureus. Ga-MOFs exhibited a bactericidal effect against these three pathogens by disrupting biofilm formation, exopolysaccharide production, and bacterial membrane integrity. In addition, we found that 1 μg/mL of Ga-MOFs was not cytotoxic to human oral epithelial cell (HOEC) lines and it significantly reduced the adhesion of the three pathogens to HOEC. Ga-MOFs protect macrophages from excessive oxidative stress by scavenging excess intracellular reactive oxygen species and upregulating antioxidant gene levels, thereby enhancing cellular antioxidant defense. In addition, Ga-MOFs can promote the transformation of macrophages from the proinflammatory phenotype to the anti-inflammatory phenotype, thereby protecting oral health. Herein, novel Ga-MOF materials were chemically synthesized for therapeutic applications in oral infections, which provides new ideas for the development of novel nonantibiotic drugs to accelerate patient recovery.
Collapse
Affiliation(s)
- Gongyuan Song
- Shijiazhuang Stomatology Hospital, Shijiazhuang, 050000, China
| | - Min Li
- Handan Stomatology Hospital, Handan, 056000, China
| | - Bing Zhou
- Cangzhou People's Hospital, Cangzhou, 061000, China
| | - Hongguang Qi
- Gucheng County Hospital of Hebei Provence, 253800, China
| | - Jie Guo
- Department of Stomatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| |
Collapse
|
7
|
Liu M, You J, Zhang Y, Zhang L, Quni S, Wang H, Zhou Y. Glucose-Responsive Self-Healing Bilayer Drug Microneedles Promote Diabetic Wound Healing Via a Trojan-Horse Strategy. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38690969 DOI: 10.1021/acsami.4c03050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Chronic nonhealing wounds are serious complications of diabetes with a high morbidity, and they can lead to disability or death. Conventional drug therapy is ineffective for diabetic wound healing because of the complex environment of diabetic wounds and the depth of drug penetration. Here, we developed a self-healing, dual-layer, drug-carrying microneedle (SDDMN) for diabetic wound healing. This SDDMN can realize transdermal drug delivery and broad-spectrum sterilization without drug resistance and meets the multiple needs of the diabetic wound healing process. Quaternary ammonium chitosan cografted with dihydrocaffeic acid (Da) and l-arginine and oxidized hyaluronic acid-dopamine are the main parts of the self-healing hydrogel patch. Methacrylated poly(vinyl alcohol) (methacrylated PVA) and phenylboronic acid (PBA) were used as the main part of the MN, and gallium porphyrin modified with 3-amino-1,2 propanediol (POGa) and insulin were encapsulated at its tip. Under hyperglycaemic conditions, the PBA moiety in the MN reversibly formed a glucose-boronic acid complex that promoted the rapid release of POGa and insulin. POGa is disguised as hemoglobin through a Trojan-horse strategy, which is then taken up by bacteria, allowing it to target bacteria and infected lesions. Based on the synergistic properties of these components, SDDMN-POGa patches exhibited an excellent biocompatibility, slow drug release, and antimicrobial properties. Thus, these patches provide a potential therapeutic approach for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Manxuan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, P. R. China
| | - Jiaqian You
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, P. R. China
| | - Yidi Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, P. R. China
| | - Lu Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, P. R. China
| | - Sezhen Quni
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, P. R. China
| | - Hanchi Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, P. R. China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun 130021, P. R. China
| |
Collapse
|
8
|
Sun W, Sun J, Ding Q, Qi M, Zhou J, Shi Y, Liu J, Won M, Sun X, Bai X, Dong B, Kim JS, Wang L. Breaking Iron Homeostasis: Iron Capturing Nanocomposites for Combating Bacterial Biofilm. Angew Chem Int Ed Engl 2024; 63:e202319690. [PMID: 38320965 DOI: 10.1002/anie.202319690] [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: 12/19/2023] [Revised: 01/22/2024] [Accepted: 02/06/2024] [Indexed: 02/08/2024]
Abstract
Given the scarcity of novel antibiotics, the eradication of bacterial biofilm infections poses formidable challenges. Upon bacterial infection, the host restricts Fe ions, which are crucial for bacterial growth and maintenance. Having coevolved with the host, bacteria developed adaptive pathways like the hemin-uptake system to avoid iron deficiency. Inspired by this, we propose a novel strategy, termed iron nutritional immunity therapy (INIT), utilizing Ga-CT@P nanocomposites constructed with gallium, copper-doped tetrakis (4-carboxyphenyl) porphyrin (TCPP) metal-organic framework, and polyamine-amine polymer dots, to target bacterial iron intakes and starve them. Owing to the similarity between iron/hemin and gallium/TCPP, gallium-incorporated porphyrin potentially deceives bacteria into uptaking gallium ions and concurrently extracts iron ions from the surrounding bacteria milieu through the porphyrin ring. This strategy orchestrates a "give and take" approach for Ga3+/Fe3+ exchange. Simultaneously, polymer dots can impede bacterial iron metabolism and serve as real-time fluorescent iron-sensing probes to continuously monitor dynamic iron restriction status. INIT based on Ga-CT@P nanocomposites induced long-term iron starvation, which affected iron-sulfur cluster biogenesis and carbohydrate metabolism, ultimately facilitating biofilm eradication and tissue regeneration. Therefore, this study presents an innovative antibacterial strategy from a nutritional perspective that sheds light on refractory bacterial infection treatment and its future clinical application.
Collapse
Affiliation(s)
- Wenyue Sun
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Jiao Sun
- Department of Cell Biology, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, China
| | - Qihang Ding
- Department of Chemistry, Korea University, Seoul, 02841, Republic of, Korea
| | - Manlin Qi
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Jing Zhou
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Yujia Shi
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Jia Liu
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Miae Won
- Department of Chemistry, Korea University, Seoul, 02841, Republic of, Korea
- TheranoChem Incorporation, Seoul, 02856, Republic of, Korea
| | - Xiaolin Sun
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Xue Bai
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, Republic of, Korea
- TheranoChem Incorporation, Seoul, 02856, Republic of, Korea
| | - Lin Wang
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| |
Collapse
|
9
|
Duffin RN, Andrews PC. Enhanced antibacterial activity of dimethyl gallium quinolinolates toward drug-resistant Klebsiella pneumoniae in low iron environments. J Inorg Biochem 2023; 249:112371. [PMID: 37738699 DOI: 10.1016/j.jinorgbio.2023.112371] [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: 07/08/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023]
Abstract
A series of dimethylgallium quinolinolate [GaMe2L] (L = 5-chloroquinolinolate, 5, 7-dichloroquinolinolate, 5, 7-dibromoquinolinolate or 5, 7-doiodoquinolinolate) complexes, shown previously to be active toward the Leishmania parasite, have been studied for their antibacterial activity toward a reference and drug resistant strain of Klebsiella pneumoniae (KP). The assays were conducted in standard iron-rich LB media and in the iron depleted RPMI and RPMI-HS media to better understand the effect of Fe concentration on the activity of the Ga complexes. In LB broth the parent quinolinols and the gallium complexes were inactive up to the highest concentration tested, 100 μM. In the more physiologically relevant 'iron-poor' RPMI-HS media the quinolonols remained inactive, however, the gallium complexes showed exceptional activity in the range 48-195 nM. Only in RPMI without any added HS did both the quinolinols and the gallium complexes show good activity. The significant differences in activity across the various media types suggest that the unnaturally high iron content of conventional LB media may provide false negative results for potentially potent Ga therapeutics. A protein binding assay on the organometallic gallium complexes showed a much slower uptake of Ga by Fe-binding proteins than is typically observed for gallium salts. This indicates that their greater lipophilicity and greater hydrolytic stability could account for their increased biological activity in RPMI-HS media.
Collapse
Affiliation(s)
- Rebekah N Duffin
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia
| | - Philip C Andrews
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia.
| |
Collapse
|
10
|
Chen T, Du H, Zhou H, He Y, Yang J, Li C, Wei C, Yu D, Wan H. Yinhuapinggan granule ameliorates lung injury caused by multidrug-resistant Acinetobacter baumannii via inhibiting NF-κB/NLRP3 pathway. Heliyon 2023; 9:e21871. [PMID: 38027639 PMCID: PMC10661428 DOI: 10.1016/j.heliyon.2023.e21871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/13/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Yinhuapinggan granule (YHPG) is a traditional Chinese medicine prescription with rich clinical experience for the treatment of colds and coughs. The aim of this study is to investigate the protective effect of YHPG on multidrug-resistant (MDR) Acinetobacter baumannii (A. baumannii) infection in vivo and its potential anti-inflammatory mechanism. BALB/c mice were intranasally inoculated with MDR A. baumannii strain to establish the pneumonia infection model, and received intraperitoneally cyclophosphamide to form immunosuppression before attack. YHPG (6, 12 and 18 g/kg) was administered by gavage once a day for 3 consecutive days after infection. The protective effect of YHPG was evaluated by lung index, spleen index, thymus index, pathological changes of lung tissue and inflammatory factors (IL-1β, IL-6 and TNF-α) in serum. The expression of key targets of NF-κB/NLRP3 signaling pathway in vivo was analyzed by immunohistochemistry, immunofluorescence, reverse transcription quantitative PCR (RT-qPCR) and Western blot. The results showed that YHPG improved the lung index and its inhibition rate, immune organ indexes and lung pathological changes in infected mice, and significantly reduced IL-1β, IL-6 and TNF-α levels in serum. In addition, YHPG significantly down-regulated the mRNA and protein expression of NF-κB p65, NLRP3, ASC, Caspase-1, TNF-α, IL-6 and IL-1β in mice lung tissue. The results of the current study demonstrated that YHPG has significant protective effects on mice infected with MDR A.baumannii, which may be related to the regulation of inflammatory factors and NF-κB/NLRP3 signaling pathway, indicating that YHPG has a wide range of clinical application value and provides a theoretical basis for its treatment of MDR A.baumannii infection.
Collapse
Affiliation(s)
- Tianhang Chen
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Haixia Du
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yu He
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chang Li
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chenxing Wei
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Daojun Yu
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
11
|
Rivera M. Mobilization of iron stored in bacterioferritin, a new target for perturbing iron homeostasis and developing antibacterial and antibiofilm molecules. J Inorg Biochem 2023; 247:112306. [PMID: 37451083 DOI: 10.1016/j.jinorgbio.2023.112306] [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: 05/05/2023] [Revised: 06/08/2023] [Accepted: 06/24/2023] [Indexed: 07/18/2023]
Abstract
Antibiotic resistance is a global public health threat. The care of chronic infections is complicated by bacterial biofilms. Biofilm embedded cells can be up to 1000-fold more tolerant to antibiotic treatment than planktonic cells. Antibiotic tolerance is a condition which does not involve mutation and enables bacteria to survive in the presence of antibiotics. The antibiotic tolerance of biofilm-cells often renders antibiotics ineffective, even against strains that do not carry resistance-impairing mutations. This review discusses bacterial iron homeostasis and the strategies being developed to target this bacterial vulnerability, with emphasis on a recently proposed approach which aims at targeting the iron storage protein bacterioferritin (Bfr) and its physiological partner, the ferredoxin Bfd. Bfr regulates cytosolic iron concentrations by oxidizing Fe2+ and storing Fe3+ in its internal cavity, and by forming a complex with Bfd to reduce Fe3+ in the internal cavity and release Fe2+ to the cytosol. Blocking the Bfr-Bfd complex in P. aeruginosa cells causes an irreversible accumulation of Fe3+ in BfrB and simultaneous cytosolic iron depletion, which leads to impaired biofilm maintenance and biofilm cell death. Recently discovered small molecule inhibitors of the Bfr-Bfd complex, which bind Bfr at the Bfd binding site, inhibit iron mobilization, and elicit biofilm cell death.
Collapse
Affiliation(s)
- Mario Rivera
- Department of Chemistry, Louisiana State University, 232 Choppin Hall, Baton Rouge, LA 70803, USA.
| |
Collapse
|
12
|
Erinmez M, Zer Y. Effects of deferoxamine on intrinsic colistin resistance of Proteus mirabilis. Exp Ther Med 2023; 26:459. [PMID: 37614438 PMCID: PMC10443054 DOI: 10.3892/etm.2023.12158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 07/20/2023] [Indexed: 08/25/2023] Open
Abstract
Proteus mirabilis is a common pathogen, which is responsible for urinary tract infections. Iron is a critical element necessary for both humans and pathogens to maintain their biological functions, and iron limitation via chelator agents may be useful in the treatment of infections. The present study aimed to investigate the synergistic interactions between the iron chelator agent deferoxamine (DFO) and the antibacterial drug colistin. The minimum inhibitory concentration (MIC) values of DFO and colistin for P. mirabilis isolates were determined by broth microdilution. The checkerboard technique was used to examine the potential synergy between DFO and colistin. Furthermore, time-kill assays were used for the confirmation of synergy detected by the checkerboard assay, as well as for determining bacteriostatic and bactericidal interactions throughout a 24-h period. As expected, all P. mirabilis isolates were resistant to colistin. DFO did not inhibit P. mirabilis growth when used alone, even at very high doses (10 µg ml-1). Notably, when in combination with DFO, the MIC values of colistin were markedly reduced, and the checkerboard assay results showed synergy between colistin and DFO for all isolates. In addition, in time-kill assays, colistin + DFO exhibited synergistic activity against all strains at most time intervals and concentrations tested. Colistin + DFO showed bactericidal activity at colistin concentrations of 1xMIC and 2xMIC, although a degree of re-growth was observed in one of the strains at 12-24 h. These findings indicated that DFO has the potential for use as an adjunct to colistin through iron sequestration, thus providing synergistic activity to an antibiotic that would not normally be considered a treatment option against P. mirabilis. In vivo experiments in the future may provide useful information on the efficacy of DFO/colistin since these models effectively reflect physiological parameters.
Collapse
Affiliation(s)
- Mehmet Erinmez
- Department of Medical Microbiology, Gaziantep University School of Medicine, 27310 Gaziantep, Turkey
| | - Yasemin Zer
- Department of Medical Microbiology, Gaziantep University School of Medicine, 27310 Gaziantep, Turkey
| |
Collapse
|
13
|
Feizi S, Awad M, Nepal R, Cooksley CM, Psaltis AJ, Wormald PJ, Vreugde S. Deferiprone-gallium-protoporphyrin (IX): A promising treatment modality against Mycobacterium abscessus. Tuberculosis (Edinb) 2023; 142:102390. [PMID: 37506532 DOI: 10.1016/j.tube.2023.102390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
Non-Tuberculous Mycobacterial Pulmonary Disease (NTM-PD) caused by Mycobacterium abscessus is a frequent complication in patients with cystic fibrosis (CF) that worsens lung function over time. Currently, there is no cure for NTM-PD, hence new therapies are urgently required. Disrupting bacterial iron uptake pathways using gallium-protoporphyrin (IX) (GaPP), a heme analog, has been proposed as a novel antibacterial approach to tackle multi-drug resistant M. abscessus. However, the antibacterial activity of GaPP has been tested only in iron-deficient media, which cannot accurately mirror the potential activity in vivo. Herein, we investigated the potential synergistic activity between GaPP and the iron-chelating agent deferiprone (Def) in regular media against M. abscessus-infected macrophages. The safety of the treatment was assessed in vitro using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay in Nuli-1 and THP-1 cell lines. Def-GaPP had synergistic activity against M. abscessus-infected macrophages where 10 mM-12.5 mg/L of Def-GaPP reduced the viability by up to 0.9 log10. Furthermore, Def-GaPP showed no cytotoxicity to Nuli-1 and THP-1 cell lines at the effective antibacterial concentrations (10 mM-12.5 mg/L) of Def- GaPP. These data encourage future investigation of Def-GaPP as a novel antimicrobial against NTM-PD.
Collapse
Affiliation(s)
- Sholeh Feizi
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, Australia; The University of Adelaide, Adelaide, Australia
| | - Muhammed Awad
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, Australia; The University of Adelaide, Adelaide, Australia; Faculty of Pharmacy, Department of Pharmaceutical Analytical Chemistry, Al-Azhar University, Assiut, Egypt
| | - Roshan Nepal
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, Australia; The University of Adelaide, Adelaide, Australia
| | - Clare M Cooksley
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, Australia; The University of Adelaide, Adelaide, Australia
| | - Alkis J Psaltis
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, Australia; The University of Adelaide, Adelaide, Australia
| | - Peter-John Wormald
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, Australia; The University of Adelaide, Adelaide, Australia
| | - Sarah Vreugde
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, Australia; The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
14
|
More O'Ferrall L, Piatek M, Twamley B, Kavanagh K, O'Connor C, Griffith DM. Synthesis, characterisation and antibacterial activity of novel Ga(III) polypyridyl catecholate complexes. Dalton Trans 2023; 52:11958-11964. [PMID: 37577980 DOI: 10.1039/d3dt01761c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Ga(III) polypyridyl catecholate complexes of type [Ga(bipy)2(O,O)](NO3) or [Ga(phen)2(O,O)](NO3) respectively were readily synthesised on reaction of Ga(NO3)3 in methanol with 1 equivalent of catecholate ligand (2,3-DHBA, 3,4-DHBA, 2,3,4-THBA or CafA) and then 2 equivalents of either bipy or phen. The complexes were characterised in full including by X-ray crystallography, which established that the catecholate ligands coordinate the Ga(III) centres in a bidentate manner via the two deprotonated hydroxy groups. All Ga(III) complexes exhibited good in vitro antibacterial activity against the Gram-negative pathogenic bacteria Escherichia coli, Klebsiella pneumoniae and Pseudomonas aeruginosa. The complexes were inactive against the Gram-positive pathogenic bacteria Staphylococcus aureus including against a methicillin-resistant Staphylococcus aureus strain (MRSA). [Ga(bipy)2(2,3-DHBA-2H)](NO3)·1.5H2O (1) was shown to be non toxic in vivo in larvae of Galleria mellonella at doses up to 2000 μg mL-1 and to offer protection at doses of 100 and 250 μg mL-1 at 48 and 96 h to larvae infected with P. aeruginosa.
Collapse
Affiliation(s)
- Lewis More O'Ferrall
- School of Food Science & Environmental Health, Technological University Dublin, Dublin 7, Ireland
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland.
| | - Magdalena Piatek
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland.
- Department of Biology, Maynooth University, Ireland
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, University of Dublin, Dublin 2, Ireland
| | - Kevin Kavanagh
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland.
- Department of Biology, Maynooth University, Ireland
| | - Christine O'Connor
- School of Food Science & Environmental Health, Technological University Dublin, Dublin 7, Ireland
| | - Darren M Griffith
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland.
- Department of Chemistry, RCSI, 123 St. Stephens Green, Dublin 2, Ireland
| |
Collapse
|
15
|
Guo M, Tian P, Li Q, Meng B, Ding Y, Liu Y, Li Y, Yu L, Li J. Gallium Nitrate Enhances Antimicrobial Activity of Colistin against Klebsiella pneumoniae by Inducing Reactive Oxygen Species Accumulation. Microbiol Spectr 2023; 11:e0033423. [PMID: 37272820 PMCID: PMC10434156 DOI: 10.1128/spectrum.00334-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/08/2023] [Indexed: 06/06/2023] Open
Abstract
Klebsiella pneumoniae, a pathogen of critical clinical concern, urgently demands effective therapeutic options owing to its drug resistance. Polymyxins are increasingly regarded as a last-line therapeutic option for the treatment of multidrug-resistant (MDR) Gram-negative bacterial infections. However, polymyxin resistance in K. pneumoniae is an emerging issue. Here, we report that gallium nitrate (GaNt), an antimicrobial candidate, exhibits a potentiating effect on colistin against MDR K. pneumoniae clinical isolates. To further confirm this, we investigated the efficacy of combined GaNt and colistin in vitro using spot dilution and rapid time-kill assays and growth curve inhibition tests and in vivo using a murine lung infection model. The results showed that GaNt significantly increased the antimicrobial activity of colistin, especially in the iron-limiting media. Mechanistic studies demonstrated that bacterial antioxidant activity was repressed by GaNt, as revealed by RNA sequencing (RNA-seq), leading to intracellular accumulation of reactive oxygen species (ROS) in K. pneumoniae, which was enhanced in the presence of colistin. Therefore, oxidative stress induced by GaNt and colistin augments the colistin-mediated killing of wild-type cells, which can be abolished by dimethyl sulfoxide (DMSO), an effective ROS scavenger. Collectively, our study indicates that GaNt has a notable impact on the antimicrobial activity of colistin against K. pneumoniae, revealing the potential of GaNt as a novel colistin adjuvant to improve the treatment outcomes of bacterial infections. IMPORTANCE This study aimed to determine the antimicrobial activity of GaNt combined with colistin against Klebsiella pneumoniae in vitro and in vivo. Our results suggest that by combining GaNt with colistin, antioxidant activity was suppressed and reactive oxygen species accumulation was induced in bacterial cells, enhancing antimicrobial activity against K. pneumoniae. We found that GaNt functioned as an antibiotic adjuvant when combined with colistin by inhibiting the growth of multidrug-resistant K. pneumoniae. Our study provides insight into the use of an adjuvant to boost the antibiotic potential of colistin for treating infections caused by multidrug-resistant K. pneumoniae.
Collapse
Affiliation(s)
- Mingjuan Guo
- Department of Infectious Disease, The Chaohu Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ping Tian
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qingqing Li
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bao Meng
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuting Ding
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanyan Liu
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui, China
| | - Yasheng Li
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui, China
| | - Liang Yu
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui, China
| | - Jiabin Li
- Department of Infectious Disease, The Chaohu Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
16
|
Venkateswaran P, Vasudevan S, David H, Shaktivel A, Shanmugam K, Neelakantan P, Solomon AP. Revisiting ESKAPE Pathogens: virulence, resistance, and combating strategies focusing on quorum sensing. Front Cell Infect Microbiol 2023; 13:1159798. [PMID: 37457962 PMCID: PMC10339816 DOI: 10.3389/fcimb.2023.1159798] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
The human-bacterial association is long-known and well-established in terms of both augmentations of human health and attenuation. However, the growing incidents of nosocomial infections caused by the ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter sp.) call for a much deeper understanding of these organisms. Adopting a holistic approach that includes the science of infection and the recent advancements in preventing and treating infections is imperative in designing novel intervention strategies against ESKAPE pathogens. In this regard, this review captures the ingenious strategies commissioned by these master players, which are teamed up against the defenses of the human team, that are equally, if not more, versatile and potent through an analogy. We have taken a basketball match as our analogy, dividing the human and bacterial species into two teams playing with the ball of health. Through this analogy, we make the concept of infectious biology more accessible.
Collapse
Affiliation(s)
- Parvathy Venkateswaran
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Sahana Vasudevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Adityan Shaktivel
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Karthik Shanmugam
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Prasanna Neelakantan
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
17
|
Sánchez-Jiménez A, Marcos-Torres FJ, Llamas MA. Mechanisms of iron homeostasis in Pseudomonas aeruginosa and emerging therapeutics directed to disrupt this vital process. Microb Biotechnol 2023. [PMID: 36857468 DOI: 10.1111/1751-7915.14241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/13/2023] [Indexed: 03/03/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen able to infect any human tissue. One of the reasons for its high adaptability and colonization of host tissues is its capacity of maintaining iron homeostasis through a wide array of iron acquisition and removal mechanisms. Due to their ability to cause life-threatening acute and chronic infections, especially among cystic fibrosis and immunocompromised patients, and their propensity to acquire resistance to many antibiotics, the World Health Organization (WHO) has encouraged the scientific community to find new strategies to eradicate this pathogen. Several recent strategies to battle P. aeruginosa focus on targeting iron homeostasis mechanisms, turning its greatest advantage into an exploitable weak point. In this review, we discuss the different mechanisms used by P. aeruginosa to maintain iron homeostasis and the strategies being developed to fight this pathogen by blocking these mechanisms. Among others, the use of iron chelators and mimics, as well as disruption of siderophore production and uptake, have shown promising results in reducing viability and/or virulence of this pathogen. The so-called 'Trojan-horse' strategy taking advantage of the siderophore uptake systems is emerging as an efficient method to improve delivery of antibiotics into the bacterial cells. Moreover, siderophore transporters are considered promising targets for the developing of P. aeruginosa vaccines.
Collapse
Affiliation(s)
- Ana Sánchez-Jiménez
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín-Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Francisco J Marcos-Torres
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín-Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - María A Llamas
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín-Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
18
|
Jeong GJ, Khan F, Khan S, Tabassum N, Mehta S, Kim YM. Pseudomonas aeruginosa virulence attenuation by inhibiting siderophore functions. Appl Microbiol Biotechnol 2023; 107:1019-1038. [PMID: 36633626 DOI: 10.1007/s00253-022-12347-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023]
Abstract
Pseudmonas aeruginosa is a Gram-negative bacterium known to be ubiquitous and recognized as one of the leading causes of infections such as respiratory, urinary tract, burns, cystic fibrosis, and in immunocompromised individuals. Failure of antimicrobial therapy has been documented to be attributable due to the development of various resistance mechanisms, with a proclivity to develop additional resistance mechanisms rapidly. P. aeruginosa virulence attenuation is an alternate technique for disrupting pathogenesis without impacting growth. The iron-scavenging siderophores (pyoverdine and pyochelin) generated by P. aeruginosa have various properties like scavenging iron, biofilm formation, quorum sensing, increasing virulence, and toxicity to the host. As a result, developing an antivirulence strategy, specifically inhibiting the P. aeruginosa siderophore, has been a promising therapeutic option to limit their infection. Several natural, synthetic compounds and nanoparticles have been identified as potent inhibitors of siderophore production/biosynthesis, function, and transport system. The current review discussed pyoverdine and pyochelin's synthesis and transport system in P. aeruginosa. Furthermore, it is also focused on the role of several natural and synthetic compounds in reducing P. aeruginosa virulence by inhibiting siderophore synthesis, function, and transport. The underlying mechanism involved in inhibiting the siderophore by natural and synthetic compounds has also been explained. KEY POINTS: • Pseudomonas aeruginosa is an opportunistic pathogen linked to chronic respiratory, urinary tract, and burns infections, as well as cystic fibrosis and immunocompromised patients. • P. aeruginosa produces two virulent siderophores forms: pyoverdine and pyochelin, which help it to survive in iron-deficient environments. • The inhibition of siderophore production, transport, and activity using natural and synthesized drugs has been described as a potential strategy for controlling P. aeruginosa infection.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea. .,Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea.
| | - Sohail Khan
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, Uttar Pradesh, 201309, India
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea.,Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Sonu Mehta
- Anthem Biosciences Private Limited, Bommasandra, Bangalore, Karnataka, 56009, India
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea. .,Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea. .,Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
19
|
Shamkani F, Barzi SM, Badmasti F, Chiani M, Mirabzadeh E, Zafari M, Shafiei M. Enhanced anti-biofilm activity of the minocycline-and-gallium-nitrate using niosome wrapping against Acinetobacter baumannii in C57/BL6 mouse pneumonia model. Int Immunopharmacol 2023; 115:109551. [PMID: 36621329 DOI: 10.1016/j.intimp.2022.109551] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/24/2022] [Accepted: 12/02/2022] [Indexed: 01/09/2023]
Abstract
Acinetobacter baumannii is a worldwide health issue in terms of its high antibiotic resistance and ability to form biofilms. Nanoparticles (NPs) with high biocompatibility, high penetrating ability, and low medication dose can successfully treat the antibiotic-resistant infections. In this research, the anti-biofilm activity of niosomes containing minocycline and gallium nitrate (GaN) against A. baumannii biofilm was determined. In order to improve their anti-biofilm properties, minocycline and GaN were encapsulated in niosomes as biocompatible drug carriers. The niosomes' size, zeta potential, shape, stability, drug entrapment efficacy, drug release pattern and antibacterial activity were assessed. Several clinical samples were isolated from the lungs of patients hospitalized at Loghman hospital, Tehran, Iran. The biofilm formation of most lethal clinical isolates of A. baumannii was analyzed. The pneumonia model was generated by intranasally administering A. baumannii suspension to anesthetized mice whose immune systems was compromised twice by cyclophosphamide. Lung infection of the mouse with A. baumannii was confirmed using PCR. After treatment, the lungs were excised under sterile conditions and stained with hematoxylin and eosin (H&E) to determine histological symptoms, inflammation and intercellular secretions. The niosomes contained minocycline and GaN had an average size of 230 nm and a zeta potential of -40 mV, respectively. The percentage of drug entrapment and delayed drug release was both high in niosomal formulations. Niosomes containing minocycline and GaN dispersed 1, 3 and 5 day old biofilms. The mice given the combination of two compounds required less time to be treated than the animals given the single medication (minocycline). The minocycline& GaN-loaded niosomes could be considered as promising candidates to treat the infections caused by A. baumannii biofilm.
Collapse
Affiliation(s)
- Farnaz Shamkani
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Mohsen Chiani
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Esmat Mirabzadeh
- Department of Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Zafari
- Department of Biology, The University of Akron, Akron, OH 44325, United States
| | - Morvarid Shafiei
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
20
|
Yang N, Shi N, Yao Z, Liu H, Guo W. Gallium-modified gelatin nanoparticles loaded with quercetin promote skin wound healing via the regulation of bacterial proliferation and macrophage polarization. Front Bioeng Biotechnol 2023; 11:1124944. [PMID: 36777248 PMCID: PMC9908762 DOI: 10.3389/fbioe.2023.1124944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/13/2023] [Indexed: 01/27/2023] Open
Abstract
Background: Wound healing is a complicated process involving multiple cell components and can help the re-establishment of the skin's barrier function. Previous studies have pointed out that bacterial infection and sustained inflammatory reactions are the main causes of the delay of wound closure and scar formation during wound healing. The effect of current approaches for scar-free wound repair still faces many challenges, and alternative therapeutic methods are urgently needed to be established. Methods: The basic characteristics of the new-designed nanoparticles were clarified through the characterization of the material. The biocompatibility of the nanoparticles, as well as its effect on fibroblast function, anti-bacterial capacity, inflammation suppressive role, and the underlying mechanism were further verified by a panel of biochemical assays in vitro. Ultimately, pre-clinical rat model was employed to testify its role in wound healing and scar formation in vivo. Results: Firstly, gallium-modified gelatin nanoparticles loaded with quercetin was successfully established, displaying good biocompatibility and facilitative effect on fibroblast function. In addition, the nanoparticles showed prominent anti-bacterial and inflammation-suppressive effects. What's more important, the nanoparticles could also induce the polarization of macrophages from M1 to M2 phenotype to exert its inflammatory inhibitory role through TGF-β/Smad signaling pathway. Ultimately, in vivo experiment showed that the nanoparticles could effectively promote wound repair and inhibit scar formation during the process of wound healing. Conclusion: Taken together, the new nanoparticles have good anti-bacterial and anti-scar formation effects and great potential in the field of skin wound repair, which provides a promising therapeutic strategy for wound treatment.
Collapse
Affiliation(s)
- Ning Yang
- Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Nianyuan Shi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi’an Jiaotong University School of Life Science and Technology, Xi’an, China
| | - Zhou Yao
- Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hang Liu
- State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an, China,*Correspondence: Hang Liu, ; Weinan Guo,
| | - Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China,*Correspondence: Hang Liu, ; Weinan Guo,
| |
Collapse
|
21
|
Mlakić M, Odak I, Faraho I, Bosnar M, Banjanac M, Lasić Z, Marinić Ž, Barić D, Škorić I. Synthesis, Photochemistry, Computational Study and Potential Application of New Styryl-Thiophene and Naphtho-Thiophene Benzylamines. Int J Mol Sci 2022; 24:ijms24010610. [PMID: 36614053 PMCID: PMC9820070 DOI: 10.3390/ijms24010610] [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: 11/28/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022] Open
Abstract
In this research, the synthesis, photochemistry, and computational study of new cis- and trans-isomers of amino-thienostilbenes is performed to test the efficiency of their production and acid resistance, and to investigate their electronic structure, photoreactivity, photophysical characteristics, and potential biological activity. The electronic structure and conformations of synthesized thienostilbene amines and their photocyclization products are examined computationally, along with molecular modeling of amines possessing two thiophene rings that showed inhibitory potential toward cholinesterases. New amino-styryl thiophenes, with favorable photophysical properties and proven acid resistance, represent model compounds for their water-soluble ammonium salts as potential styryl optical dyes. The comparison with organic dyes possessing a trans-aminostilbene subunit as the scaffold shows that the newly synthesized trans-aminostilbenes have very similar absorbance wavelengths. Furthermore, their functionalized cis-isomers and photocyclization products are good candidates for cholinesterase inhibitors because of the structural similarity of the molecular skeleton to some already proven bioactive derivatives.
Collapse
Affiliation(s)
- Milena Mlakić
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia
| | - Ilijana Odak
- Department of Chemistry, Faculty of Science and Education, University of Mostar, Matice Hrvatske bb, 88000 Mostar, Bosnia and Herzegovina
| | - Ivan Faraho
- Pharmacology In Vitro, Selvita Ltd., Prilaz Baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Martina Bosnar
- Pharmacology In Vitro, Selvita Ltd., Prilaz Baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Mihailo Banjanac
- Pharmacology In Vitro, Selvita Ltd., Prilaz Baruna Filipovića 29, HR-10000 Zagreb, Croatia
| | - Zlata Lasić
- Teva api Analytical R&D, Pliva, Prilaz Baruna Filipovića 25, HR-10000 Zagreb, Croatia
| | - Željko Marinić
- NMR Center, Ruđer Bošković Institute, Bijenička Cesta 54, HR-10000 Zagreb, Croatia
| | - Danijela Barić
- Group for Computational Life Sciences, Division of Physical Chemistry, Ruđer Bošković Institute, Bijenička Cesta 54, HR-10000 Zagreb, Croatia
- Correspondence: (D.B.); (I.Š.); Tel.: +385-1-4571-385 (D.B.); +385-1-4597-241 (I.Š.)
| | - Irena Škorić
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia
- Correspondence: (D.B.); (I.Š.); Tel.: +385-1-4571-385 (D.B.); +385-1-4597-241 (I.Š.)
| |
Collapse
|
22
|
Dong J, Wang W, Zhou W, Zhang S, Li M, Li N, Pan G, Zhang X, Bai J, Zhu C. Immunomodulatory biomaterials for implant-associated infections: from conventional to advanced therapeutic strategies. Biomater Res 2022; 26:72. [PMID: 36471454 PMCID: PMC9721013 DOI: 10.1186/s40824-022-00326-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/19/2022] [Indexed: 12/11/2022] Open
Abstract
Implant-associated infection (IAI) is increasingly emerging as a serious threat with the massive application of biomaterials. Bacteria attached to the surface of implants are often difficult to remove and exhibit high resistance to bactericides. In the quest for novel antimicrobial strategies, conventional antimicrobial materials often fail to exert their function because they tend to focus on direct bactericidal activity while neglecting the modulation of immune systems. The inflammatory response induced by host immune cells was thought to be a detrimental force impeding wound healing. However, the immune system has recently received increasing attention as a vital player in the host's defense against infection. Anti-infective strategies based on the modulation of host immune defenses are emerging as a field of interest. This review explains the importance of the immune system in combating infections and describes current advanced immune-enhanced anti-infection strategies. First, the characteristics of traditional/conventional implant biomaterials and the reasons for the difficulty of bacterial clearance in IAI were reviewed. Second, the importance of immune cells in the battle against bacteria is elucidated. Then, we discuss how to design biomaterials that activate the defense function of immune cells to enhance the antimicrobial potential. Based on the key premise of restoring proper host-protective immunity, varying advanced immune-enhanced antimicrobial strategies were discussed. Finally, current issues and perspectives in this field were offered. This review will provide scientific guidance to enhance the development of advanced anti-infective biomaterials.
Collapse
Affiliation(s)
- Jiale Dong
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Wenzhi Wang
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Wei Zhou
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Siming Zhang
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Meng Li
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China ,grid.263761.70000 0001 0198 0694Medical College, Soochow University, 215006 Suzhou, Jiangsu P. R. China
| | - Ning Li
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Guoqing Pan
- grid.440785.a0000 0001 0743 511XInstitute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 212013 Zhenjiang, China
| | - Xianzuo Zhang
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Jiaxiang Bai
- grid.263761.70000 0001 0198 0694Medical College, Soochow University, 215006 Suzhou, Jiangsu P. R. China
| | - Chen Zhu
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| |
Collapse
|
23
|
Al-Madboly LA. A Novel Triple Combination To Combat Serious Infections with Carbapenem-Resistant Acinetobacter baumannii in a Mouse Pneumonia Model. Microbiol Spectr 2022; 10:e0271021. [PMID: 35975993 PMCID: PMC9603289 DOI: 10.1128/spectrum.02710-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/17/2022] [Indexed: 01/04/2023] Open
Abstract
The ongoing crisis of antimicrobial resistance demands novel combinations between antimicrobials and nonantimicrobials to manage infections caused by highly resistant pathogens. This study aimed to evaluate the effect of combining sodium ascorbate and/or apo-transferrin with imipenem, forming double and triple combinations, against 20 multiple-carbapenemase-producing Acinetobacter baumannii strains using the checkerboard test, time-kill assay, and disc diffusion test. The results of the checkerboard assay revealed that all double combinations showed indifference, while only triple combination recorded a synergistic effect (fractional inhibitory concentration index [FICI] < 0.8) in 95% the test isolates. Moreover, the MIC of imipenem (MICimp) was strongly reduced (up to 128-fold reduction) after treatment with the triple combination against highly resistant isolates and reached the susceptible range. The time-kill assay revealed that the triple combination led to a 4-log10 reduction in the CFU at 8 h compared with the initial bacterial count, and no viable count was recorded at 10 h. The mouse pneumonia model showed restoration of lung function and structure, with mild to moderate residual inflammation and moderately congested vessels observed 8 h following administration of the triple rescue therapy. Additionally, normal lungs with normal patent alveoli were detected 72 h following treatment. Accordingly, sodium ascorbate and apo-transferrin are promising adjunct biological agents with the potential to restore the effectiveness of critically essential antibiotics like imipenem, commonly used for the treatment of A. baumannii infections. IMPORTANCE Combination therapy provides a perspective to threat multidrug-resistant (MDR) strains. The present study sheds light on a novel and effective triple combination against carbapenem-resistant A. baumannii. Our in vitro results showed that combining imipenem with apo-transferrin and sodium ascorbate yielded synergism in 95% of test isolates, and this was associated with a marked reduction in imipenem MIC, shifting it below the breakpoint. Furthermore, a bactericidal effect was recorded, with no viable count detected at 10 h. An in vivo murine model of pneumonia was induced to mimic human disease. The triple combination therapy restored lung function and structure, with mild to moderate residual inflammation and moderately congested vessels observed 8 h following the initiation of therapy. Therefore, our findings suggest novel insights about a promising new combination therapy against highly resistant carbapenemase-producing A. baumannii to restore the effectiveness of imipenem.
Collapse
Affiliation(s)
- Lamiaa A. Al-Madboly
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
24
|
Vargová Z, Rendošová M, Saksová S, Gyepes R, Vilková M. Complexing properties of 2-pyridylphoshonate and 2-pyridylsulfonate ligands for Zn 2+ and Ag + central atoms. J COORD CHEM 2022. [DOI: 10.1080/00958972.2022.2127095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Zuzana Vargová
- Department of Inorganic Chemistry, Faculty of Science, P. J. Šafárik University, Košice, Slovak Republic
| | - Michaela Rendošová
- Department of Inorganic Chemistry, Faculty of Science, P. J. Šafárik University, Košice, Slovak Republic
| | - Silvia Saksová
- Department of Inorganic Chemistry, Faculty of Science, P. J. Šafárik University, Košice, Slovak Republic
| | - Róbert Gyepes
- Department of Inorganic Chemistry, Faculty of Science, Charles University, Praha, Czech Republic
| | - Mária Vilková
- NMR Laboratory, Faculty of Science, P. J. Šafárik University, Košice, Slovak Republic
| |
Collapse
|
25
|
Ma R, Hu X, Zhang X, Wang W, Sun J, Su Z, Zhu C. Strategies to prevent, curb and eliminate biofilm formation based on the characteristics of various periods in one biofilm life cycle. Front Cell Infect Microbiol 2022; 12:1003033. [PMID: 36211965 PMCID: PMC9534288 DOI: 10.3389/fcimb.2022.1003033] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Biofilms are colonies of bacteria embedded inside a complicated self-generating intercellular. The formation and scatter of a biofilm is an extremely complex and progressive process in constant cycles. Once formed, it can protect the inside bacteria to exist and reproduce under hostile conditions by establishing tolerance and resistance to antibiotics as well as immunological responses. In this article, we reviewed a series of innovative studies focused on inhibiting the development of biofilm and summarized a range of corresponding therapeutic methods for biological evolving stages of biofilm. Traditionally, there are four stages in the biofilm formation, while we systematize the therapeutic strategies into three main periods precisely:(i) period of preventing biofilm formation: interfering the colony effect, mass transport, chemical bonds and signaling pathway of plankton in the initial adhesion stage; (ii) period of curbing biofilm formation:targeting several pivotal molecules, for instance, polysaccharides, proteins, and extracellular DNA (eDNA) via polysaccharide hydrolases, proteases, and DNases respectively in the second stage before developing into irreversible biofilm; (iii) period of eliminating biofilm formation: applying novel multifunctional composite drugs or nanoparticle materials cooperated with ultrasonic (US), photodynamic, photothermal and even immune therapy, such as adaptive immune activated by stimulated dendritic cells (DCs), neutrophils and even immunological memory aroused by plasmocytes. The multitargeted or combinational therapies aim to prevent it from developing to the stage of maturation and dispersion and eliminate biofilms and planktonic bacteria simultaneously.
Collapse
Affiliation(s)
| | | | | | | | | | - Zheng Su
- *Correspondence: Chen Zhu, ; Zheng Su,
| | - Chen Zhu
- *Correspondence: Chen Zhu, ; Zheng Su,
| |
Collapse
|
26
|
Ali IAA, Cheung GS, Neelakantan P. Transition Metals and
Enterococcus faecalis
: Homeostasis, Virulence and Perspectives. Mol Oral Microbiol 2022; 37:276-291. [DOI: 10.1111/omi.12391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Islam A. A. Ali
- Department of Endodontics Faculty of Dentistry Mansoura University Mansoura Egypt
| | - Gary S.P. Cheung
- Discipline of Endodontology Division of Restorative Dental Sciences Faculty of Dentistry The University of Hong Kong Hong Kong SAR
| | - Prasanna Neelakantan
- Discipline of Endodontology Division of Restorative Dental Sciences Faculty of Dentistry The University of Hong Kong Hong Kong SAR
| |
Collapse
|
27
|
Repac Antić D, Parčina M, Gobin I, Petković Didović M. Chelation in Antibacterial Drugs: From Nitroxoline to Cefiderocol and Beyond. Antibiotics (Basel) 2022; 11:1105. [PMID: 36009974 PMCID: PMC9405089 DOI: 10.3390/antibiotics11081105] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
In the era of escalating antimicrobial resistance, the need for antibacterial drugs with novel or improved modes of action (MOAs) is a health concern of utmost importance. Adding or improving the chelating abilities of existing drugs or finding new, nature-inspired chelating agents seems to be one of the major ways to ensure progress. This review article provides insight into the modes of action of antibacterial agents, class by class, through the perspective of chelation. We covered a wide scope of antibacterials, from a century-old quintessential chelating agent nitroxoline, currently unearthed due to its newly discovered anticancer and antibiofilm activities, over the commonly used antibacterial classes, to new cephalosporin cefiderocol and a potential future class of tetramates. We show the impressive spectrum of roles that chelation plays in antibacterial MOAs. This, by itself, demonstrates the importance of understanding the fundamental chemistry behind such complex processes.
Collapse
Affiliation(s)
- Davorka Repac Antić
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Department of Clinical Microbiology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Marijo Parčina
- Institute of Medical Microbiology, Immunology and Parasitology, Bonn University Hospital, 53127 Bonn, Germany
| | - Ivana Gobin
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Mirna Petković Didović
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
28
|
Ammendola S, Battistoni A. New Insights into the Role of Metals in Host-Pathogen Interactions. Int J Mol Sci 2022; 23:ijms23126483. [PMID: 35742927 PMCID: PMC9224429 DOI: 10.3390/ijms23126483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022] Open
Abstract
Almost eighty years have passed since the publication of the studies by Arthur Schade and Leona Caroline, which we can consider as the first investigations that began to disclose the importance of metals in host-pathogen interactions [...].
Collapse
Affiliation(s)
- Serena Ammendola
- Correspondence: (S.A.); (A.B.); Tel.: +39-06-0672594368 (S.A.); +39-06-7259-4372 (A.B.)
| | - Andrea Battistoni
- Correspondence: (S.A.); (A.B.); Tel.: +39-06-0672594368 (S.A.); +39-06-7259-4372 (A.B.)
| |
Collapse
|
29
|
Law SKK, Tan HS. The Role of Quorum Sensing, Biofilm Formation, and Iron Acquisition as Key Virulence Mechanisms in Acinetobacter baumannii and the Corresponding Anti-virulence Strategies. Microbiol Res 2022; 260:127032. [DOI: 10.1016/j.micres.2022.127032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/15/2022]
|
30
|
Visaggio D, Frangipani E, Hijazi S, Pirolo M, Leoni L, Rampioni G, Imperi F, Bernstein L, Sorrentino R, Ungaro F, Visca P. Variable Susceptibility to Gallium Compounds of Major Cystic Fibrosis Pathogens. ACS Infect Dis 2022; 8:78-85. [PMID: 34965085 PMCID: PMC8762661 DOI: 10.1021/acsinfecdis.1c00409] [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] [Indexed: 01/31/2023]
Abstract
![]()
The decreasing efficacy
of existing antibiotics against pulmonary
pathogens that affect cystic fibrosis (CF) patients calls for the
development of novel antimicrobials. Iron uptake and metabolism are
vital processes for bacteria, hence potential therapeutic targets.
Gallium [Ga(III)] is a ferric iron-mimetic that inhibits bacterial
growth by disrupting iron uptake and metabolism. In this work we evaluate
the efficacy of three Ga(III) compounds, namely, Ga(NO3)3, (GaN), Ga(III)-maltolate (GaM), and Ga(III)-protoporphyrin
IX (GaPPIX), against a collection of CF pathogens using both reference
media and media mimicking biological fluids. All CF pathogens, except Streptococcus pneumoniae, were susceptible to at
least one Ga(III) compound. Notably, Mycobacterium
abscessus and Stenotrophomonas maltophilia were susceptible to all Ga(III) compounds. Achromobacter
xylosoxidans, Burkholderia cepacia complex, and Pseudomonas aeruginosa were more susceptible to GaN and GaM, whereas Staphylococcus
aureus and Haemophilus influenzae were more sensitive to GaPPIX. The results of this study support
the development of Ga(III)-based therapy as a broad-spectrum strategy
to treat CF lung infections.
Collapse
Affiliation(s)
- Daniela Visaggio
- Department of Science, Roma Tre University, 00146 Rome, Italy
- Santa Lucia Fundation IRCCS, 00179 Rome, Italy
| | - Emanuela Frangipani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Sarah Hijazi
- Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Mattia Pirolo
- Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Livia Leoni
- Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Giordano Rampioni
- Department of Science, Roma Tre University, 00146 Rome, Italy
- Santa Lucia Fundation IRCCS, 00179 Rome, Italy
| | - Francesco Imperi
- Department of Science, Roma Tre University, 00146 Rome, Italy
- Santa Lucia Fundation IRCCS, 00179 Rome, Italy
| | | | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80138 Naples, Italy
| | - Francesca Ungaro
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Paolo Visca
- Department of Science, Roma Tre University, 00146 Rome, Italy
- Santa Lucia Fundation IRCCS, 00179 Rome, Italy
| |
Collapse
|
31
|
Iron Homeostasis in Pseudomonas aeruginosa: Targeting Iron Acquisition and Storage as an Antimicrobial Strategy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1386:29-68. [DOI: 10.1007/978-3-031-08491-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
32
|
Salmonella Typhimurium and Pseudomonas aeruginosa Respond Differently to the Fe Chelator Deferiprone and to Some Novel Deferiprone Derivatives. Int J Mol Sci 2021; 22:ijms221910217. [PMID: 34638558 PMCID: PMC8508819 DOI: 10.3390/ijms221910217] [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: 07/15/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
The ability to obtain Fe is critical for pathogens to multiply in their host. For this reason, there is significant interest in the identification of compounds that might interfere with Fe management in bacteria. Here we have tested the response of two Gram-negative pathogens, Salmonella enterica serovar Typhimurium (STM) and Pseudomonas aeruginosa (PAO1), to deferiprone (DFP), a chelating agent already in use for the treatment of thalassemia, and to some DFP derivatives designed to increase its lipophilicity. Our results indicate that DFP effectively inhibits the growth of PAO1, but not STM. Similarly, Fe-dependent genes of the two microorganisms respond differently to this agent. DFP is, however, capable of inhibiting an STM strain unable to synthesize enterochelin, while its effect on PAO1 is not related to the capability to produce siderophores. Using a fluorescent derivative of DFP we have shown that this chelator can penetrate very quickly into PAO1, but not into STM, suggesting that a selective receptor exists in Pseudomonas. Some of the tested derivatives have shown a greater ability to interfere with Fe homeostasis in STM compared to DFP, whereas most, although not all, were less active than DFP against PAO1, possibly due to interference of the added chemical tails with the receptor-mediated recognition process. The results reported in this work indicate that DFP can have different effects on distinct microorganisms, but that it is possible to obtain derivatives with a broader antimicrobial action.
Collapse
|
33
|
Nasser A, Dallal MMS, Jahanbakhshi S, Azimi T, Nikouei L. Staphylococcus aureus: biofilm formation and strategies against it. Curr Pharm Biotechnol 2021; 23:664-678. [PMID: 34238148 DOI: 10.2174/1389201022666210708171123] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/09/2021] [Accepted: 05/31/2021] [Indexed: 11/22/2022]
Abstract
The formation of Staphylococcus aureus biofilm causes significant infections in the human body. Biofilm forms through the aggregation of bacterial species and brings about many complications. It mediates drug resistance and persistence and facilitates the recurrence of infection at the end of antimicrobial therapy. Biofilm formation goes through a series of steps to complete, and any interference in these steps can disrupt its formation. Such interference may occur at any stage of biofilm production, including attachment, monolayer formation, and accumulation. Interfering agents can act as quorum sensing inhibitors and interfere in the functionality of quorum sensing receptors, attachment inhibitors and affect the cell hydrophobicity. Among these inhibiting strategies, attachment inhibitors could serve as the best agents against biofilm formation. If pathogens abort the attachment, the following stages of biofilm formation, e.g., accumulation and dispersion, will fail to materialize. Inhibition at this stage leads to suppression of virulence factors and invasion. One of the best-known inhibitors is a chelator that collects metal, Fe+, Zn+, and magnesium critical for biofilm formation. These influential factors in the binding and formation of biofilm are investigated, and the coping strategy is discussed. This review examines the stages of biofilm formation and determines what factors interfere in the continuity of these steps. Finally, the inhibition strategies are investigated, reviewed, and discussed. Keywords: Biofilm, Staphylococcus, Biofilm inhibitor, Dispersion, Antibiofilm agent, EPS, PIA.
Collapse
Affiliation(s)
- Ahmad Nasser
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shiva Jahanbakhshi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Taher Azimi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Nikouei
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Vinuesa V, Cruces R, Nonnoi F, McConnell MJ. Inhibition of LpxC Increases the Activity of Iron Chelators and Gallium Nitrate in Multidrug-Resistant Acinetobacter baumannii. Antibiotics (Basel) 2021; 10:antibiotics10050609. [PMID: 34065605 PMCID: PMC8160660 DOI: 10.3390/antibiotics10050609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 11/17/2022] Open
Abstract
Infections caused by multidrug-resistant Acinetobacter baumannii would benefit from the development of novel treatment approaches. Compounds that interfere with bacterial iron metabolism, such as iron chelators and gallium nitrate, have previously been shown to have antimicrobial activity against A. baumannii. In this study, we characterize the effect of LpxC inhibitors on the antimicrobial activity of previously characterized iron chelators, 2,2′-bipyridyl (BIP) and deferiprone (DFP), and gallium nitrate (Ga(NO3)3) against A. baumannii reference strains and multidrug-resistant clinical isolates. The LpxC inhibitor LpxC-2 was synergistic with BIP for 30% of strains tested (FICI values: 0.38–1.02), whereas inhibition with LpxC-4 was synergistic with BIP for 60% of strains tested (FICI values: 0.09–0.75). In time–kill assays, combinations of BIP with both LpxC inhibitors demonstrated synergistic activity, with a more than 3 log10 reduction in bacterial counts compared to BIP alone. LpxC-2 was synergistic with Ga(NO3)3 for 50% of strains tested (FICI values: 0.27–1.0), whereas LpxC-4 was synergistic with Ga(NO3)3 for all strains tested (FICI values: 0.08–≤0.50). In time–kill assays, combinations of Ga(NO3)3 with LpxC-2 and LpxC-4 decreased the growth of both strains compared to each compound separately; however, only the combination with LpxC-4 met the defined criteria for synergy. These results identify a novel synergy between two antimicrobial classes against A. baumannii strains.
Collapse
Affiliation(s)
- Víctor Vinuesa
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28220 Madrid, Spain; (V.V.); (R.C.); (F.N.)
| | - Raquel Cruces
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28220 Madrid, Spain; (V.V.); (R.C.); (F.N.)
| | - Francesca Nonnoi
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28220 Madrid, Spain; (V.V.); (R.C.); (F.N.)
| | - Michael J. McConnell
- Vaxdyn S.L., Avenida Manuel Siurot s/n., 41010 Seville, Spain
- Correspondence: ; Tel.: +34-918-223-869
| |
Collapse
|