1
|
Aggarwal M, Patra A, Awasthi I, George A, Gagneja S, Gupta V, Capalash N, Sharma P. Drug repurposing against antibiotic resistant bacterial pathogens. Eur J Med Chem 2024; 279:116833. [PMID: 39243454 DOI: 10.1016/j.ejmech.2024.116833] [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: 05/06/2024] [Revised: 08/22/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
The growing prevalence of MDR and XDR bacterial pathogens is posing a critical threat to global health. Traditional antibiotic development paths have encountered significant challenges and are drying up thus necessitating innovative approaches. Drug repurposing, which involves identifying new therapeutic applications for existing drugs, offers a promising alternative to combat resistant pathogens. By leveraging pre-existing safety and efficacy data, drug repurposing accelerates the development of new antimicrobial therapy regimes. This review explores the potential of repurposing existing FDA approved drugs against the ESKAPE and other clinically relevant bacterial pathogens and delves into the identification of suitable drug candidates, their mechanisms of action, and the potential for combination therapies. It also describes clinical trials and patent protection of repurposed drugs, offering perspectives on this evolving realm of therapeutic interventions against drug resistance.
Collapse
Affiliation(s)
- Manya Aggarwal
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Anushree Patra
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Ishita Awasthi
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Annu George
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Simran Gagneja
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Varsha Gupta
- Department of Microbiology, Government Multi-speciality hospital, Sector 16, Chandigarh, India
| | - Neena Capalash
- Department of Biotechnology, Panjab University, Chandigarh, India
| | - Prince Sharma
- Departmen of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
2
|
Guo C, Wang KKA, Nolan EM. Investigation of Siderophore-Platinum(IV) Conjugates Reveals Differing Antibacterial Activity and DNA Damage Depending on the Platinum Cargo. ACS Infect Dis 2024; 10:1250-1266. [PMID: 38436588 DOI: 10.1021/acsinfecdis.3c00686] [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] [Indexed: 03/05/2024]
Abstract
The growing threat of bacterial infections coupled with the dwindling arsenal of effective antibiotics has heightened the urgency for innovative strategies to combat bacterial pathogens, particularly Gram-negative strains, which pose a significant challenge due to their outer membrane permeability barrier. In this study, we repurpose clinically approved anticancer agents as targeted antibacterials. We report two new siderophore-platinum(IV) conjugates, both of which consist of an oxaliplatin-based Pt(IV) prodrug (oxPt(IV)) conjugated to enterobactin (Ent), a triscatecholate siderophore employed by Enterobacteriaceae for iron acquisition. We demonstrate that l/d-Ent-oxPt(IV) (l/d-EOP) are selectively delivered into the Escherichia coli cytoplasm, achieving targeted antibacterial activity, causing filamentous morphology, and leading to enhanced Pt uptake by bacterial cells but reduced Pt uptake by human cells. d-EOP exhibits enhanced potency compared to oxaliplatin and l-EOP, primarily attributed to the intrinsic antibacterial activity of its non-native siderophore moiety. To further elucidate the antibacterial activity of Ent-Pt(IV) conjugates, we probed DNA damage caused by l/d-EOP and the previously reported cisplatin-based conjugates l/d-Ent-Pt(IV) (l/d-EP). A comparative analysis of these four conjugates reveals a correlation between antibacterial activity and the ability to induce DNA damage. This work expands the scope of Pt cargos targeted to the cytoplasm of Gram-negative bacteria via Ent conjugation, provides insight into the cellular consequences of Ent-Pt(IV) conjugates in E. coli, and furthers our understanding of the potential of Pt-based therapeutics for antibacterial applications.
Collapse
Affiliation(s)
- Chuchu Guo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Kwo-Kwang A Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Elizabeth M Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
3
|
Chen ER, Wozniak RAF. Reimagining the Past: A Future for Antibiotic Drug Discovery in Ophthalmology. Cornea 2024; 43:1-5. [PMID: 37702607 DOI: 10.1097/ico.0000000000003391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/09/2023] [Indexed: 09/14/2023]
Abstract
ABSTRACT Antibiotic resistance has emerged as a critical threat for the treatment of bacterial ocular infections. To address the critical need for novel therapeutics, antibiotic drug repurposing holds significant promise. As such, examples of existing FDA-approved drugs currently under development for new applications, novel combinations, and improved delivery systems are discussed.
Collapse
Affiliation(s)
- Eric R Chen
- Department of Ophthalmology, Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY
| | | |
Collapse
|
4
|
Savelyeva IO, Zhdanova KA, Gradova MA, Gradov OV, Bragina NA. Cationic Porphyrins as Antimicrobial and Antiviral Agents in Photodynamic Therapy. Curr Issues Mol Biol 2023; 45:9793-9822. [PMID: 38132458 PMCID: PMC10741785 DOI: 10.3390/cimb45120612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023] Open
Abstract
Antimicrobial photodynamic therapy (APDT) has received a great deal of attention due to its unique ability to kill all currently known classes of microorganisms. To date, infectious diseases caused by bacteria and viruses are one of the main sources of high mortality, mass epidemics and global pandemics among humans. Every year, the emergence of three to four previously unknown species of viruses dangerous to humans is recorded, totaling more than 2/3 of all newly discovered human pathogens. The emergence of bacteria with multidrug resistance leads to the rapid obsolescence of antibiotics and the need to create new types of antibiotics. From this point of view, photodynamic inactivation of viruses and bacteria is of particular interest. This review summarizes the most relevant mechanisms of antiviral and antibacterial action of APDT, molecular targets and correlation between the structure of cationic porphyrins and their photodynamic activity.
Collapse
Affiliation(s)
- Inga O. Savelyeva
- Institute of Fine Chemical Technology, MIREA—Russian Technological University, Vernadsky Prospect 86, Moscow 119571, Russia; (I.O.S.); (K.A.Z.); (N.A.B.)
| | - Kseniya A. Zhdanova
- Institute of Fine Chemical Technology, MIREA—Russian Technological University, Vernadsky Prospect 86, Moscow 119571, Russia; (I.O.S.); (K.A.Z.); (N.A.B.)
| | - Margarita A. Gradova
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Kosygin Street 4, Moscow 119991, Russia;
| | - Oleg V. Gradov
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Kosygin Street 4, Moscow 119991, Russia;
| | - Natal’ya A. Bragina
- Institute of Fine Chemical Technology, MIREA—Russian Technological University, Vernadsky Prospect 86, Moscow 119571, Russia; (I.O.S.); (K.A.Z.); (N.A.B.)
| |
Collapse
|
5
|
Kacsir I, Sipos A, Major E, Bajusz N, Bényei A, Buglyó P, Somsák L, Kardos G, Bai P, Bokor É. Half-Sandwich Type Platinum-Group Metal Complexes of C-Glucosaminyl Azines: Synthesis and Antineoplastic and Antimicrobial Activities. Molecules 2023; 28:molecules28073058. [PMID: 37049820 PMCID: PMC10096180 DOI: 10.3390/molecules28073058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/01/2023] Open
Abstract
While platinum-based compounds such as cisplatin form the backbone of chemotherapy, the use of these compounds is limited by resistance and toxicity, driving the development of novel complexes with cytostatic properties. In this study, we synthesized a set of half-sandwich complexes of platinum-group metal ions (Ru(II), Os(II), Ir(III) and Rh(III)) with an N,N-bidentate ligand comprising a C-glucosaminyl group and a heterocycle, such as pyridine, pyridazine, pyrimidine, pyrazine or quinoline. The sugar-containing ligands themselves are unknown compounds and were obtained by nucleophilic additions of lithiated heterocycles to O-perbenzylated 2-nitro-glucal. Reduction of the adducts and, where necessary, subsequent protecting group manipulations furnished the above C-glucosaminyl heterocycles in their O-perbenzylated, O-perbenzoylated and O-unprotected forms. The derived complexes were tested on A2780 ovarian cancer cells. Pyridine, pyrazine and pyridazine-containing complexes proved to be cytostatic and cytotoxic on A2780 cells, while pyrimidine and quinoline derivatives were inactive. The best complexes contained pyridine as the heterocycle. The metal ion with polyhapto arene/arenyl moiety also impacted on the biological activity of the complexes. Ruthenium complexes with p-cymene and iridium complexes with Cp* had the best performance in ovarian cancer cells, followed by osmium complexes with p-cymene and rhodium complexes with Cp*. Finally, the chemical nature of the protective groups on the hydroxyl groups of the carbohydrate moiety were also key determinants of bioactivity; in particular, O-benzyl groups were superior to O-benzoyl groups. The IC50 values of the complexes were in the low micromolar range, and, importantly, the complexes were less active against primary, untransformed human dermal fibroblasts; however, the anticipated therapeutic window is narrow. The bioactive complexes exerted cytostasis on a set of carcinomas such as cell models of glioblastoma, as well as breast and pancreatic cancers. Furthermore, the same complexes exhibited bacteriostatic properties against multiresistant Gram-positive Staphylococcus aureus and Enterococcus clinical isolates in the low micromolar range.
Collapse
Affiliation(s)
- István Kacsir
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
- Doctoral School of Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., H-4032 Debrecen, Hungary
- The Hungarian Academy of Sciences, Center of Excellence, Hungary
- MTA-DE Cell Biology and Signaling Research Group ELKH, H-4032 Debrecen, Hungary
| | - Evelin Major
- Department of Metagenomics, University of Debrecen, H-4032 Debrecen, Hungary
| | - Nikolett Bajusz
- Department of Metagenomics, University of Debrecen, H-4032 Debrecen, Hungary
| | - Attila Bényei
- Department of Physical Chemistry, Faculty of Sciences and Technology, University of Debrecen, Egyetem Tér 1., H-4032 Debrecen, Hungary
| | - Péter Buglyó
- Department of Inorganic & Analytical Chemistry, Faculty of Sciences and Technology, University of Debrecen, Egyetem Tér 1., H-4032 Debrecen, Hungary
| | - László Somsák
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
| | - Gábor Kardos
- Department of Metagenomics, University of Debrecen, H-4032 Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., H-4032 Debrecen, Hungary
- The Hungarian Academy of Sciences, Center of Excellence, Hungary
- MTA-DE Cell Biology and Signaling Research Group ELKH, H-4032 Debrecen, Hungary
- NKFIH-DE Lendület Laboratory of Cellular Metabolism, H-4032 Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
- Correspondence: (P.B.); (É.B.); Tel.: +36-524-123-45 (P.B.); +36-525-129-00 (ext. 22474) (É.B.)
| | - Éva Bokor
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
- Correspondence: (P.B.); (É.B.); Tel.: +36-524-123-45 (P.B.); +36-525-129-00 (ext. 22474) (É.B.)
| |
Collapse
|
6
|
Kacsir I, Sipos A, Kiss T, Major E, Bajusz N, Tóth E, Buglyó P, Somsák L, Kardos G, Bai P, Bokor É. Half sandwich-type osmium, ruthenium, iridium and rhodium complexes with bidentate glycosyl heterocyclic ligands induce cytostasis in platinum-resistant ovarian cancer cells and bacteriostasis in Gram-positive multiresistant bacteria. Front Chem 2023; 11:1086267. [PMID: 36793764 PMCID: PMC9923724 DOI: 10.3389/fchem.2023.1086267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/05/2023] [Indexed: 01/31/2023] Open
Abstract
The toxicity of and resistance to platinum complexes as cisplatin, oxaliplatin or carboplatin calls for the replacement of these therapeutic agents in clinical settings. We have previously identified a set of half sandwich-type osmium, ruthenium and iridium complexes with bidentate glycosyl heterocyclic ligands exerting specific cytostatic activity on cancer cells but not on non-transformed primary cells. The apolar nature of the complexes, conferred by large, apolar benzoyl protective groups on the hydroxyl groups of the carbohydrate moiety, was the main molecular feature to induce cytostasis. We exchanged the benzoyl protective groups to straight chain alkanoyl groups with varying length (3 to 7 carbon units) that increased the IC50 value as compared to the benzoyl-protected complexes and rendered the complexes toxic. These results suggest a need for aromatic groups in the molecule. The pyridine moiety of the bidentate ligand was exchanged for a quinoline group to enlarge the apolar surface of the molecule. This modification decreased the IC50 value of the complexes. The complexes containing [(η6-p-cymene)Ru(II)], [(η6-p-cymene)Os(II)] or [(η5-Cp*)Ir(III)] were biologically active unlike the complex containing [(η5-Cp*)Rh(III)]. The complexes with cytostatic activity were active on ovarian cancer (A2780, ID8), pancreatic adenocarcinoma (Capan2), sarcoma (Saos) and lymphoma cell lines (L428), but not on primary dermal fibroblasts and their activity was dependent on reactive oxygen species production. Importantly, these complexes were cytostatic on cisplatin-resistant A2780 ovarian cancer cells with similar IC50 values as on cisplatin-sensitive A2780 cells. In addition, the quinoline-containing Ru and Os complexes and the short chain alkanoyl-modified complexes (C3 and C4) proved to be bacteriostatic in multiresistant Gram-positive Enterococcus and Staphylococcus aureus isolates. Hereby, we identified a set of complexes with submicromolar to low micromolar inhibitory constants against a wide range of cancer cells, including platinum resistant cells and against multiresistant Gram-positive bacteria.
Collapse
Affiliation(s)
- István Kacsir
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
- Doctoral School of Chemistry, University of Debrecen, Debrecen, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tímea Kiss
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
| | - Evelin Major
- Department of Metagenomics, University of Debrecen, Debrecen, Hungary
| | - Nikolett Bajusz
- Department of Metagenomics, University of Debrecen, Debrecen, Hungary
| | - Emese Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Buglyó
- Department of Inorganic and Analytical Chemistry, Faculty of Sciences and Technology, University of Debrecen, Debrecen, Hungary
| | - László Somsák
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
| | - Gábor Kardos
- Department of Metagenomics, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- NKFIH-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group ELKH, Debrecen, Hungary
| | - Éva Bokor
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
7
|
Bari AK, Belalekar TS, Poojary A, Rohra S. Combination drug strategies for biofilm eradication using synthetic and natural agents in KAPE pathogens. Front Cell Infect Microbiol 2023; 13:1155699. [PMID: 37139491 PMCID: PMC10149696 DOI: 10.3389/fcimb.2023.1155699] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/29/2023] [Indexed: 05/05/2023] Open
Abstract
Antibiotic resistance is a global threat caused by factors such as overuse of antibiotics, lack of awareness, development of biofilms etc. World Health Organization released a list of global priority pathogens which consisted of 12 species of bacteria categorized as expressing critical, high and medium resistance. Several Gram-negative and Gram-positive species are known to cause wide varieties of infections and have become multidrug or extremely drug resistant. Pathogens causing infections associated with invasive medical devices are biofilm producers and hence their treatment becomes difficult due to a structurally stable matrix which prevents antibiotics from penetrating the biofilm and thereby showing its effects. Factors contributing to tolerance are inhibition of penetration, restricted growth and activation of biofilm genes. Combination drug therapies has also shown potential to eradicate biofilm infections. A combination of inhaled Fosfomycin/tobramycin antibiotic strategy has been effective against Gram-negative as well as Gram positive organisms. Along with antibiotics, use of natural or synthetic adjuvants shows promising effects to treat biofilm infections. Fluroquinolone activity on biofilms is disrupted by low oxygen tension in the matrix, a strategy known as hyperbaric oxygen treatment that can enhance efficacy of antibiotics if well optimized. Adjuvants such as Ethylenediaminetetraacetic acid (EDTA), Sodium Dodecyl Sulphate (SDS) and chlorhexidine act by killing non-growing microbial cells aggregated on the inner layer of the biofilm. This review aims to list down current combination therapies used against Gram-negative and Gram-positive biofilm forming pathogens and brief about comparison of combination drugs and their efficacies.
Collapse
|
8
|
Clerici DJ, Hahn da Silveira C, Iglesias BA, Christ Vianna Santos R. The first evidence of antibiofilm action of Proteus mirabilis with tetra-cationic porphyrins containing cisplatin by antimicrobial photodynamic therapy. Microb Pathog 2023; 174:105859. [PMID: 36403712 DOI: 10.1016/j.micpath.2022.105859] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022]
Abstract
Biofilms are responsible for up to 80% of antimicrobial-resistant nosocomial infections. Most of these infections are associated with medical devices such as urinary catheters, and in this context, it is estimated that 90-100% of patients who undergo long-term catheterization develop infections. Proteus mirabilis, the most prevalent microorganism, is responsible for 20-45% of these infections. Thus, this study aimed to evaluate, for the first time, the antimicrobial and antibiofilm effects of cationic porphyrins on P. mirabilis. Neutral porphyrins 3-H2TPyP and 4-H2TpyP and tetra-cationic derivatives 3-cis-PtTPyP and 4-cis-PtTPyP were evaluated in broth microdilution tests to determine the minimum inhibitory and bactericidal concentrations. Time-kill curves, checkerboard test, reactive oxygen species (ROS) scavenger assays, conventional biofilm formation, and biofilm assay with catheters were also performed. The microdilution tests showed greater efficacy against P. mirabilis when 3-cis-PtTPyP was exposed to white-light conditions; this also occurred when the microbial time-kill curve was performed at 0, 2, 6, and 12 h. The radical superoxide species was possibly responsible for photoinactivation in the ROS scavenger assays. In biofilm assays (conventional and catheter), 3-cis-PtTPyP obtained better results when irradiated with a white-light source. In the checkerboard assay, the same compound showed no differences when tested in association with ciprofloxacin hydrochloride. Our findings lead us to conclude that antimicrobial photodynamic therapy and cationic porphyrins obtained positive results and are promising alternatives to treat P. mirabilis biofilms.
Collapse
Affiliation(s)
- Dariane Jornada Clerici
- Laboratório De Pesquisa Em Microbiologia Oral, Departamento De Microbiologia e Parasitologia, Universidade Federal De Santa Maria, Av. Roraima 1000, 97105-900, Santa Maria, RS, Brazil
| | - Carolina Hahn da Silveira
- Laboratório De Bioinorgânica e Materiais Porfirínicos, Departamento De Química, Universidade Federal De Santa Maria, UFSM, Av. Roraima 1000, 97105-900, Santa Maria, RS, Brazil
| | - Bernardo Almeida Iglesias
- Laboratório De Bioinorgânica e Materiais Porfirínicos, Departamento De Química, Universidade Federal De Santa Maria, UFSM, Av. Roraima 1000, 97105-900, Santa Maria, RS, Brazil.
| | - Roberto Christ Vianna Santos
- Laboratório De Pesquisa Em Microbiologia Oral, Departamento De Microbiologia e Parasitologia, Universidade Federal De Santa Maria, Av. Roraima 1000, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
9
|
Chowdhury N, Bagchi A. A drug repurposing endeavor to discover a multi-targeting ligand against RhlR and LasR proteins from opportunistic human pathogen Pseudomonas aeruginosa. J Mol Model 2022; 28:295. [PMID: 36064977 DOI: 10.1007/s00894-022-05301-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: 09/20/2021] [Accepted: 08/29/2022] [Indexed: 11/25/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen. It synthesizes the poison called Hydrogen Cyanide (HCN). The synthesis of HCN is mediated by the enzyme HCN synthase which is obtained from the hcnABC operon and the transcription of the hcnABC operon is mediated by three proteins LasR, RhlR, and ANR. In our previous works, we analyzed the activation process of RhlR and LasR proteins by their cognate auto-inducer ligands (N-butanoyl-L-homoserine lactone and N-(3-oxododecanoyl)-homoserine lactone respectively). In this work, we attempted to identify some multi-targeting ligands which would be able to destroy the structural integrity of both the RhlR and LasR proteins using steered MD simulations. We used the virtual screening of ligand libraries, and for that purpose, we used the NCI drug database. We selected the top 4 ligands from our virtual screening experiments. We then tried to check their relative binding affinities with the LasR and RhlR proteins in comparison to their native auto-inducer ligands. Through this work, we were able to identify 4 such ligands which were capable of binding to both the RhlR and LasR proteins in a better way than their native auto-inducer ligands. The efficacies of these ligands to actually perturb the structural integrity of RhlR and LasR proteins could be tested in wet lab. The work is the first work in the field of structure-based drug design to come up with possible multi-targeting drug-like structures against the RhlR and LasR proteins from Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- Nilkanta Chowdhury
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, Kalyani, 741235, West Bengal, India
- Department of Biotechnology, Sidho-Kanho-Birsha University, Ranchi-Purulia Road Campus, Near Sainik School, Purulia, 723104, West Bengal, India
| | - Angshuman Bagchi
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, Kalyani, 741235, West Bengal, India.
| |
Collapse
|
10
|
Jampilek J. Drug repurposing to overcome microbial resistance. Drug Discov Today 2022; 27:2028-2041. [PMID: 35561965 DOI: 10.1016/j.drudis.2022.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 12/15/2022]
Abstract
Infections are a growing global threat, and the number of resistant species of microbial pathogens is alarming. However, the rapid development of cross-resistant or multidrug-resistant strains and the development of so-called 'superbugs' are in stark contrast to the number of newly launched anti-infectives on the market. In this review, I summarize the causes of antimicrobial resistance, briefly discuss different approaches to the discovery and development of new anti-infective drugs, and focus on drug repurposing strategy, which is discussed from all possible perspectives. A comprehensive overview of drugs of other indications tested for their in vitro antimicrobial activity to support existing anti-infective therapeutics is provided, including several critical remarks on this strategy of repurposing non-antibiotics to antibacterial drugs.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia; Department of Chemical Biology, Faculty of Science, Palacky University, Slechtitelu 27, 783 71 Olomouc, Czech Republic.
| |
Collapse
|
11
|
Balázs B, Tóth Z, Kacsir I, Sipos A, Buglyó P, Somsák L, Bokor É, Kardos G, Bai P. Targeting Multiresistant Gram-Positive Bacteria by Ruthenium, Osmium, Iridium and Rhodium Half-Sandwich Type Complexes With Bidentate Monosaccharide Ligands. Front Chem 2022; 10:868234. [PMID: 35494644 PMCID: PMC9039051 DOI: 10.3389/fchem.2022.868234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Bacterial resistance to antibiotics is an ever-growing problem in heathcare. We have previously identified a set of osmium(II), ruthenium(II), iridium(III) and rhodium(III) half-sandwich type complexes with bidentate monosaccharide ligands possessing cytostatic properties against carcinoma, lymphoma and sarcoma cells with low micromolar or submicromolar IC50 values. Importantly, these complexes were not active on primary, non-transformed cells. These complexes have now been assessed as to their antimicrobial properties and found to be potent inhibitors of the growth of reference strains of Staphylococcus aureus and Enterococcus faecalis (Gram-positive species), though the compounds proved inactive on reference strains of Pseudomonas aerugonisa, Escherichia coli, Candida albicans, Candida auris and Acinetobacter baumannii (Gram-negative species and fungi). Furthermore, clinical isolates of Staphylococcus aureus and Enterococcus sp. (both multiresistant and susceptible strains) were also susceptible to the organometallic complexes in this study with similar MIC values as the reference strains. Taken together, we identified a set of osmium(II), ruthenium(II), iridium(III) and rhodium(III) half-sandwich type antineoplastic organometallic complexes which also have antimicrobial activity among Gram-positive bacteria. These compounds represent a novel class of antimicrobial agents that are not detoxified by multiresistant bacteria suggesting a potential to be used to combat multiresistant infections.
Collapse
Affiliation(s)
- Bence Balázs
- Department of Metagenomics, University of Debrecen, Debrecen, Hungary
| | - Zoltán Tóth
- Department of Metagenomics, University of Debrecen, Debrecen, Hungary
| | - István Kacsir
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
- Doctoral School of Chemistry, University of Debrecen, Debrecen, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Buglyó
- Department of Inorganic and Analytical Chemistry, Faculty of Sciences and Technology, University of Debrecen, Debrecen, Hungary
| | - László Somsák
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
| | - Éva Bokor
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
- *Correspondence: Éva Bokor, ; Gábor Kardos, ; Péter Bai,
| | - Gábor Kardos
- Department of Metagenomics, University of Debrecen, Debrecen, Hungary
- *Correspondence: Éva Bokor, ; Gábor Kardos, ; Péter Bai,
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- NKFIH-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- *Correspondence: Éva Bokor, ; Gábor Kardos, ; Péter Bai,
| |
Collapse
|
12
|
Pecoraro C, Carbone D, Deng D, Cascioferro SM, Diana P, Giovannetti E. Biofilm Formation as Valuable Target to Fight against Severe Chronic Infections. Curr Med Chem 2022; 29:4307-4310. [PMID: 34979887 DOI: 10.2174/0929867329666220103095551] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 11/22/2022]
Affiliation(s)
- Camilla Pecoraro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF) the University of Palermo, 90123 Palermo, Italy
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF) the University of Palermo, 90123 Palermo, Italy
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, 1081 HV Amsterdam, The Netherlands The Netherlands
| | - Stella Maria Cascioferro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF) the University of Palermo, 90123 Palermo, Italy
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF) the University of Palermo, 90123 Palermo, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, 56124 Pisa, Italy
| |
Collapse
|
13
|
Vyas T, Rapalli VK, Chellappan DK, Dua K, Dubey SK, Singhvi G. Bacterial biofilms associated skin disorders: Pathogenesis, advanced pharmacotherapy and nanotechnology-based drug delivery systems as a treatment approach. Life Sci 2021; 287:120148. [PMID: 34785190 DOI: 10.1016/j.lfs.2021.120148] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/30/2021] [Accepted: 11/09/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Biofilms are microcolonies of microbes that form communities with a variety of microbes, exhibit the same gene composition but differ in gene expression. Biofilm-associated infections have been in existence for a long, however, biofilm-associated skin disorders have not been investigated much. OBJECTIVES Biofilms, which are made mostly of the matrix can be thought of as communities of microbes that are more virulent and more difficult to eradicate as compared to their planktonic counterparts. Currently, several formulations are available in the market which have the potential to treat biofilm-assisted skin disorders. However, the existing pharmacotherapies are not competent enough to cure them effectively and entirely, in several cases. KEY FINDINGS Especially with the rising resistance towards antibiotics, it has become particularly challenging to ameliorate these disorders completely. The new approaches are being used to combat biofilm-associated skin disorders, some of them being photodynamic therapy, nanotherapies, and the use of novel drug delivery systems. The focus of attention, however, is nanotherapy. Micelles, solid lipid nanoparticles, quatsomes, and many others are being considered to find a better solution for the biofilm-associated skin disorders. SIGNIFICANCE This review is an attempt to give a perspective on these new approaches for treating bacterial biofilms associated with skin disorders.
Collapse
Affiliation(s)
- Taraj Vyas
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
| | | | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney (UTS), Ultimo, NSW 2007, Australia; Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, 2007 New South Wales, Australia
| | - Sunil Kumar Dubey
- Medical Research, R&D Healthcare Division, Emami Ltd, Kolkata 700056, India
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India.
| |
Collapse
|
14
|
Foletto VS, da Rosa TF, Serafin MB, Bottega A, Hörner R. Repositioning of non-antibiotic drugs as an alternative to microbial resistance: a systematic review. Int J Antimicrob Agents 2021; 58:106380. [PMID: 34166776 DOI: 10.1016/j.ijantimicag.2021.106380] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/10/2021] [Accepted: 06/10/2021] [Indexed: 01/19/2023]
Abstract
The global spread of microbial resistance coupled with high costs and slow pace in the discovery of a new antibiotic have made drug repositioning an attractive and promising alternative in the treatment of infections caused by multidrug resistant (MDR) microorganisms. The reuse involves the production of compounds with lower costs and development time, using diversified production technologies. The present systematic review aimed to present a selection of studies published in the last 20 years, which report the antimicrobial activity of non-antibiotic drugs that are candidates for repositioning, which could be used against the current microbial multidrug resistance. A search was performed in the PubMed, SciELO and Google Scholar databases using the following search strategies: [(drug repurposing) OR (drug repositioning) OR (repositioning) AND (non-antibiotic) AND (antibacterial activity) AND (antimicrobial activity)]. Overall, 112 articles were included, which explored the antimicrobial activity in antidepressants, antihypertensives, anti-inflammatories, antineoplastics, hypoglycemic agents, among other drugs. It was concluded that they have significant antimicrobial activity in vitro and in vivo, against standard strain and clinical isolates (Gram-negative and Gram-positive) and fungi. When associated with antibacterials, most of these drugs had their antibacterial activity enhanced. It was also a consensus of the studies included in this review that the presence of aromatic rings in the molecular structure contributes to antimicrobial activity. This review highlights the potential repositioning of several classes of non-antibiotic drugs as promising candidates for repositioning in the treatment of severe bacterial infections of MDR bacteria, extensively resistant (XDR) and pan-resistant (PDR) to drugs.
Collapse
Affiliation(s)
- Vitória S Foletto
- Universidade Federal de Santa Maria, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brasil
| | - Taciéli F da Rosa
- Universidade Federal de Santa Maria, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brasil
| | - Marissa B Serafin
- Universidade Federal de Santa Maria, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brasil
| | - Angelita Bottega
- Universidade Federal de Santa Maria, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brasil
| | - Rosmari Hörner
- Universidade Federal de Santa Maria, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brasil; Universidade Federal de Santa Maria, Departamento de Análises Clínicas e Toxicológicas, Santa Maria, RS, Brasil.
| |
Collapse
|
15
|
Possato B, Dalmolin LF, Pereira LM, Alves JQ, Silva RTC, Gelamo RV, Yatsuda AP, Lopez RFV, de Albuquerque S, Leite NB, Maia PIDS. Gold(III) complexes with thiosemicarbazonate ligands as potential anticancer agents: Cytotoxicity and interactions with biomolecular targets. Eur J Pharm Sci 2021; 162:105834. [PMID: 33826936 DOI: 10.1016/j.ejps.2021.105834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 01/23/2023]
Abstract
Gold(III) complexes have been studied for the past years due to their anticancer properties and great affinity to biotargets, such as enzymes and proteins, which support their pharmacological applications. Within this scope, in this work the antiproliferative activities of two Au(III)-thiosemicarbazonate complexes, [AuClL1] (1, L1: (E,Z)-N-ethyl-N'-(3-nitroso-kN)butan-2-ylidene)carbamohydrazonothioato-k2N2,S) and [Au(Hdamp)L2]Cl (2, L2: N-(N'',N''-diethylaminothiocarbonyl)-N'(N''', N'''-dimethylcarbothioamide)benzamidineto-kN,k2S and Hdamp: 2-(N,N-dimethylaminomethyl)-phenyl-C1), and their affinities to possible biological targets were investigated. Three different tumor cell lines were used to perform the cytotoxicity assays, including one cisplatin-resistant model, and the results showed lower EC50 for 1 over 2 in every case: B16F10 (4.1 μM and 15.6 μM), A431 (4.0 μM and >50 μM) and OVCAR3 (4.2 μM and 24.5 μM). However, a lower toxicity to fibroblast 3T3 cell line was observed for 2 (30.58 μM) when compared to 1 (7.17 μM), resulting in comparable therapeutic indexes. Both complexes presented strong affinity to HSA: they distorted the secondary structure of the protein, as verified by circular dichroism, but 1 additionally presented the apparent fluorescence quenching constant (Kapp) ten times greater than 2, which was probably due to the fact of 1 being able to denature HSA. The ethidium bromide displacement assay showed that neither 1 nor 2 are strong DNA intercalators, which is in agreement with what was observed through the UV-vis titration. In both cases, the 260 nm band presented hyperchromism, which can indicate ionic interactions or DNA damage. In fact, 1 was able to damage the pGEM plasmid, similarly to cisplatin, as verified by agarose gel electrophoresis and Atomic Force Microscopy. Biophysical studies in cancer cells model membranes were also performed in order to investigate the interaction of the gold complexes to lipid bilayers and revealed that the compounds interact with the membranes by exhibiting partition coefficients of 103 order of magnitude. Overall, both complexes were found to be promising candidates for the development of a future anticancer drug against low sensitive or cisplatin resistant tumors.
Collapse
Affiliation(s)
- Bruna Possato
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Luciana Falcco Dalmolin
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Luiz Miguel Pereira
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | | | - Rogerio Valentim Gelamo
- Instituto de Ciências Tecnológicas e Exatas, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | - Ana Patrícia Yatsuda
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | - Sérgio de Albuquerque
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Natália Bueno Leite
- Núcleo de Desenvolvimento de Compostos Bioativos (NDCBio), Universidade Federal do Triângulo Mineiro, Uberaba, Brazil.
| | - Pedro Ivo da Silva Maia
- Núcleo de Desenvolvimento de Compostos Bioativos (NDCBio), Universidade Federal do Triângulo Mineiro, Uberaba, Brazil.
| |
Collapse
|
16
|
Sipos A, Ujlaki G, Mikó E, Maka E, Szabó J, Uray K, Krasznai Z, Bai P. The role of the microbiome in ovarian cancer: mechanistic insights into oncobiosis and to bacterial metabolite signaling. Mol Med 2021; 27:33. [PMID: 33794773 PMCID: PMC8017782 DOI: 10.1186/s10020-021-00295-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is characterized by dysbiosis, referred to as oncobiosis in neoplastic diseases. In ovarian cancer, oncobiosis was identified in numerous compartments, including the tumor tissue itself, the upper and lower female genital tract, serum, peritoneum, and the intestines. Colonization was linked to Gram-negative bacteria with high inflammatory potential. Local inflammation probably participates in the initiation and continuation of carcinogenesis. Furthermore, local bacterial colonies in the peritoneum may facilitate metastasis formation in ovarian cancer. Vaginal infections (e.g. Neisseria gonorrhoeae or Chlamydia trachomatis) increase the risk of developing ovarian cancer. Bacterial metabolites, produced by the healthy eubiome or the oncobiome, may exert autocrine, paracrine, and hormone-like effects, as was evidenced in breast cancer or pancreas adenocarcinoma. We discuss the possible involvement of lipopolysaccharides, lysophosphatides and tryptophan metabolites, as well as, short-chain fatty acids, secondary bile acids and polyamines in the carcinogenesis of ovarian cancer. We discuss the applicability of nutrients, antibiotics, and probiotics to harness the microbiome and support ovarian cancer therapy. The oncobiome and the most likely bacterial metabolites play vital roles in mediating the effectiveness of chemotherapy. Finally, we discuss the potential of oncobiotic changes as biomarkers for the diagnosis of ovarian cancer and microbial metabolites as possible adjuvant agents in therapy.
Collapse
Affiliation(s)
- Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Gyula Ujlaki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Eszter Maka
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, Debrecen, 4032, Hungary
| | - Judit Szabó
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Karen Uray
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Zoárd Krasznai
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, Debrecen, 4032, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.
| |
Collapse
|
17
|
Lettl C, Schindele F, Testolin G, Bär A, Rehm T, Brönstrup M, Schobert R, Bilitewski U, Haas R, Fischer W. Inhibition of Type IV Secretion Activity and Growth of Helicobacter pylori by Cisplatin and Other Platinum Complexes. Front Cell Infect Microbiol 2020; 10:602958. [PMID: 33392108 PMCID: PMC7775389 DOI: 10.3389/fcimb.2020.602958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
Type IV secretion systems are protein secretion machineries that are frequently used by pathogenic bacteria to inject their virulence factors into target cells of their respective hosts. In the case of the human gastric pathogen Helicobacter pylori, the cytotoxin-associated gene (Cag) type IV secretion system is considered a major cause for severe disease, such as gastric cancer, and thus constitutes an attractive target for specific treatment options against H. pylori infections. Here, we have used a Cag type IV secretion reporter assay for screening a repurposing compound library for inhibitors targeting this system. We found that the antitumor agent cisplatin, a platinum coordination complex that kills target cells by formation of DNA crosslinks, is a potent inhibitor of the Cag type IV secretion system. Strikingly, we found that this inhibitory activity of cisplatin depends on a ligand exchange reaction which incorporates a solvent molecule (dimethylsulfoxide) into the complex, a modification which is known to be deleterious for DNA crosslinking, and for its anticancer activity. We extended our analysis to several analogous platinum complexes containing N-heterocyclic carbene, as well as DMSO or other ligands, and found varying inhibitory activities toward the Cag system which were not congruent with their DNA-binding properties, suggesting that protein interactions may cause the inhibitory effect. Inhibition experiments under varying conditions revealed effects on adherence and bacterial viability as well, and showed that the type IV secretion-inhibitory capacity of platinum complexes can be inactivated by sulfur-containing reagents and in complex bacterial growth media. Taken together, our results demonstrate DNA binding-independent inhibitory effects of cisplatin and other platinum complexes against different H. pylori processes including type IV secretion.
Collapse
Affiliation(s)
- Clara Lettl
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich Site, Munich, Germany
| | - Franziska Schindele
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich Site, Munich, Germany
| | - Giambattista Testolin
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,German Center for Infection Research (DZIF), Hannover-Braunschweig Site, Braunschweig, Germany
| | - Alexander Bär
- Organic Chemistry Laboratory, University Bayreuth, Bayreuth, Germany
| | - Tobias Rehm
- Organic Chemistry Laboratory, University Bayreuth, Bayreuth, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,German Center for Infection Research (DZIF), Hannover-Braunschweig Site, Braunschweig, Germany
| | - Rainer Schobert
- Organic Chemistry Laboratory, University Bayreuth, Bayreuth, Germany
| | - Ursula Bilitewski
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,German Center for Infection Research (DZIF), Hannover-Braunschweig Site, Braunschweig, Germany
| | - Rainer Haas
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich Site, Munich, Germany
| | - Wolfgang Fischer
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich Site, Munich, Germany
| |
Collapse
|
18
|
Hummell NA, Kirienko NV. Repurposing bioactive compounds for treating multidrug-resistant pathogens. J Med Microbiol 2020; 69:881-894. [PMID: 32163353 DOI: 10.1099/jmm.0.001172] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Introduction. Antimicrobial development is being outpaced by the rising rate of antimicrobial resistance in the developing and industrialized world. Drug repurposing, where novel antibacterial functions can be found for known molecular entities, reduces drug development costs, reduces regulatory hurdles, and increases rate of success.Aim. We sought to characterize the antimicrobial properties of five known bioactives (DMAQ-B1, carboplatin, oxaliplatin, CD437 and PSB-069) that were discovered in a high-throughput phenotypic screen for hits that extend Caenorhabditis elegans survival during exposure to Pseudomonas aeruginosa PA14.Methodology. c.f.u. assays, biofilm staining and fluorescence microscopy were used to assay the compounds' effect on various virulence determinants. Checkerboard assays were used to assess synergy between compounds and conventional antimicrobials. C. elegans-based assays were used to test whether the compounds could also rescue against Enterococcus faecalis and Staphyloccus aureus. Finally, toxicity was assessed in C. elegans and mammalian cells.Results. Four of the compounds rescued C. elegans from a second bacterial pathogen and two of them (DMAQ-B1, a naturally occurring insulin mimetic, and CD437, an agonist of the retinoic acid receptor) rescued against all three. The platinum complexes displayed increased antimicrobial activity against P. aeruginosa. Of the molecules tested, only CD437 showed slight synergy with ampicillin. The two most effective compounds, DMAQ-B1 and CD437, showed toxicity to mammalian cells.Conclusion. Although these compounds' potential for repurposing is limited by their toxicity, our results contribute to this growing field and provide a simple road map for using C. elegans for preliminary testing of known bioactive compounds with predicted antimicrobial activity.
Collapse
|
19
|
Verderosa AD, Totsika M, Fairfull-Smith KE. Bacterial Biofilm Eradication Agents: A Current Review. Front Chem 2019; 7:824. [PMID: 31850313 PMCID: PMC6893625 DOI: 10.3389/fchem.2019.00824] [Citation(s) in RCA: 313] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022] Open
Abstract
Most free-living bacteria can attach to surfaces and aggregate to grow into multicellular communities encased in extracellular polymeric substances called biofilms. Biofilms are recalcitrant to antibiotic therapy and a major cause of persistent and recurrent infections by clinically important pathogens worldwide (e.g., Pseudomonas aeruginosa, Escherichia coli, and Staphylococcus aureus). Currently, most biofilm remediation strategies involve the development of biofilm-inhibition agents, aimed at preventing the early stages of biofilm formation, or biofilm-dispersal agents, aimed at disrupting the biofilm cell community. While both strategies offer some clinical promise, neither represents a direct treatment and eradication strategy for established biofilms. Consequently, the discovery and development of biofilm eradication agents as comprehensive, stand-alone biofilm treatment options has become a fundamental area of research. Here we review our current understanding of biofilm antibiotic tolerance mechanisms and provide an overview of biofilm remediation strategies, focusing primarily on the most promising biofilm eradication agents and approaches. Many of these offer exciting prospects for the future of biofilm therapeutics for a large number of infections that are currently refractory to conventional antibiotics.
Collapse
Affiliation(s)
- Anthony D Verderosa
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,School of Chemistry, Physics, and Mechanical Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Makrina Totsika
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kathryn E Fairfull-Smith
- School of Chemistry, Physics, and Mechanical Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
20
|
Kumar R, Harilal S, Gupta SV, Jose J, Thomas Parambi DG, Uddin MS, Shah MA, Mathew B. Exploring the new horizons of drug repurposing: A vital tool for turning hard work into smart work. Eur J Med Chem 2019; 182:111602. [PMID: 31421629 PMCID: PMC7127402 DOI: 10.1016/j.ejmech.2019.111602] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
Drug discovery and development are long and financially taxing processes. On an average it takes 12-15 years and costs 1.2 billion USD for successful drug discovery and approval for clinical use. Many lead molecules are not developed further and their potential is not tapped to the fullest due to lack of resources or time constraints. In order for a drug to be approved by FDA for clinical use, it must have excellent therapeutic potential in the desired area of target with minimal toxicities as supported by both pre-clinical and clinical studies. The targeted clinical evaluations fail to explore other potential therapeutic applications of the candidate drug. Drug repurposing or repositioning is a fast and relatively cheap alternative to the lengthy and expensive de novo drug discovery and development. Drug repositioning utilizes the already available clinical trials data for toxicity and adverse effects, at the same time explores the drug's therapeutic potential for a different disease. This review addresses recent developments and future scope of drug repositioning strategy.
Collapse
Affiliation(s)
- Rajesh Kumar
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Seetha Harilal
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Sheeba Varghese Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Science, NITTE Deemed to be University, Manglore, 575018, India
| | - Della Grace Thomas Parambi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Al Jouf, 2014, Saudi Arabia
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Muhammad Ajmal Shah
- Department of Pharmacogonosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, 678557, Kerala, India.
| |
Collapse
|
21
|
Song S, Gong T, Yamasaki R, Kim J, Wood TK. Identification of a potent indigoid persister antimicrobial by screening dormant cells. Biotechnol Bioeng 2019; 116:2263-2274. [DOI: 10.1002/bit.27078] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/19/2019] [Accepted: 05/27/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Sooyeon Song
- Department of Chemical EngineeringPennsylvania State UniversityUniversity Park Pennsylvania
| | - Ting Gong
- Department of Chemical EngineeringPennsylvania State UniversityUniversity Park Pennsylvania
| | - Ryota Yamasaki
- Department of Chemical EngineeringPennsylvania State UniversityUniversity Park Pennsylvania
| | - Jun‐Seob Kim
- Infectious Disease Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeon South Korea
| | - Thomas K. Wood
- Department of Chemical EngineeringPennsylvania State UniversityUniversity Park Pennsylvania
| |
Collapse
|
22
|
Fitting Pieces into the Puzzle of Pseudomonas aeruginosa Type III Secretion System Gene Expression. J Bacteriol 2019; 201:JB.00209-19. [PMID: 31010903 DOI: 10.1128/jb.00209-19] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Type III secretion systems (T3SS) are widely distributed in Gram-negative microorganisms and critical for host-pathogen and host-symbiont interactions with plants and animals. Central features of the T3SS are a highly conserved set of secretion and translocation genes and contact dependence wherein host-pathogen interactions trigger effector protein delivery and serve as an inducing signal for T3SS gene expression. In addition to these conserved features, there are pathogen-specific properties that include a unique repertoire of effector genes and mechanisms to control T3SS gene expression. The Pseudomonas aeruginosa T3SS serves as a model system to understand transcriptional and posttranscriptional mechanisms involved in the control of T3SS gene expression. The central regulatory feature is a partner-switching system that controls the DNA-binding activity of ExsA, the primary regulator of T3SS gene expression. Superimposed upon the partner-switching mechanism are cyclic AMP and cyclic di-GMP signaling systems, two-component systems, global regulators, and RNA-binding proteins that have positive and negative effects on ExsA transcription and/or synthesis. In the present review, we discuss advances in our understanding of how these regulatory systems orchestrate the activation of T3SS gene expression in the context of acute infections and repression of the T3SS as P. aeruginosa adapts to and colonizes the cystic fibrosis airways.
Collapse
|