1
|
O'Boyle B, Yeung W, Lu JD, Katiyar S, Yaron-Barir TM, Johnson JL, Cantley LC, Kannan N. Atlas of the Bacterial Serine-Threonine Kinases expands the functional diversity of the kinome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.12.632604. [PMID: 39868133 PMCID: PMC11760699 DOI: 10.1101/2025.01.12.632604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Bacterial serine-threonine protein kinases (STKs) regulate diverse cellular processes associated with cell growth, virulence, and pathogenicity. They are evolutionarily related to the druggable eukaryotic STKs. However, an incomplete knowledge of how bacterial STKs differ from their eukaryotic counterparts and how they have diverged to regulate diverse bacterial signaling functions presents a bottleneck in targeting them for drug discovery efforts. Here, we classified over 300,000 bacterial STK sequences from the NCBI RefSeq non-redundant and UniProt protein databases into 35 canonical and seven non-canonical (pseudokinase) families based on the patterns of evolutionary constraints in the conserved catalytic domain and flanking regulatory domains. Through statistical comparisons, we identified distinguishing features of bacterial STKs, including a distinctive arginine residue in a regulatory helix (C-Helix) that dynamically couples ATP and substrate binding lobes of the kinase domain. Biochemical and peptide-library screens demonstrated that constrained residues contribute to substrate specificity and kinase activation in the Mycobacterium tuberculosis kinase PknB. Collectively, these findings open new avenues for investigating bacterial STK functions in cellular signaling and for the development of selective bacterial STK inhibitors.
Collapse
|
2
|
Cowen L, Puumala E, Nandakumar M, Yiu B, Stogios P, Strickland B, Zarnowski R, Wang X, Williams N, Savchenko A, Andes D, Robbins N, Whitesell L, Willson T. Structure-guided optimization of small molecules targeting the yeast casein kinase, Yck2, as a therapeutic strategy to combat Candida albicans. RESEARCH SQUARE 2025:rs.3.rs-5524306. [PMID: 39866870 PMCID: PMC11760248 DOI: 10.21203/rs.3.rs-5524306/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Candida albicans is the most common cause of life-threatening fungal infection in the developed world but remains a therapeutic challenge. Protein kinases have been rewarding drug targets across diverse indications but remain untapped for antifungal development. Previously, screening kinase inhibitors against C. albicans revealed a 2,3-aryl-pyrazolopyridine, GW461484A (GW), which targets casein kinase 1 (CK1) family member Yck2. Here, we report optimization of GW via two complementary approaches, synthesis of bioisosteres possessing an imidazo[1,2-a]pyridine core, and R-group substitution of GW's pyrazolo[1,5-a]pyridine core. Characterization of compounds synthesized revealed two 6-cyano derivatives with improved pharmacological properties that retained whole-cell bioactivity and selectivity for fungal Yck2 compared to human CK1α. Efficacy studies in mice indicated both analogs possess single-agent activity against C. albicans resistant to first-line echinocandin antifungals and potentiate non-curative echinocandin treatment. Results validate Yck2 as an antifungal target and encourage further development of inhibitors acting by this previously unexploited mode of action.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Noelle Williams
- The University of Texas Southwestern Medical Center at Dallas
| | | | | | | | | | | |
Collapse
|
3
|
Zhang T, Nickerson R, Zhang W, Peng X, Shang Y, Zhou Y, Luo Q, Wen G, Cheng Z. The impacts of animal agriculture on One Health-Bacterial zoonosis, antimicrobial resistance, and beyond. One Health 2024; 18:100748. [PMID: 38774301 PMCID: PMC11107239 DOI: 10.1016/j.onehlt.2024.100748] [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: 08/28/2023] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
The industrialization of animal agriculture has undoubtedly contributed to the improvement of human well-being by increasing the efficiency of food animal production. At the same time, it has also drastically impacted the natural environment and human society. The One Health initiative emphasizes the interdependency of the health of ecosystems, animals, and humans. In this paper, we discuss some of the most profound consequences of animal agriculture practices from a One Health perspective. More specifically, we focus on impacts to host-microbe interactions by elaborating on how modern animal agriculture affects zoonotic infections, specifically those of bacterial origin, and the concomitant emergence of antimicrobial resistance (AMR). A key question underlying these deeply interconnected issues is how to better prevent, monitor, and manage infections in animal agriculture. To address this, we outline approaches to mitigate the impacts of agricultural bacterial zoonoses and AMR, including the development of novel treatments as well as non-drug approaches comprising integrated surveillance programs and policy and education regarding agricultural practices and antimicrobial stewardship. Finally, we touch upon additional major environmental and health factors impacted by animal agriculture within the One Health context, including animal welfare, food security, food safety, and climate change. Charting how these issues are interwoven to comprise the complex web of animal agriculture's broad impacts on One Health will allow for the development of concerted, multidisciplinary interventions which are truly necessary to tackle these issues from a One Health perspective.
Collapse
Affiliation(s)
- Tengfei Zhang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Rhea Nickerson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Wenting Zhang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Xitian Peng
- Institute of Quality Standard and Testing Technology for Agro-Products, Hubei Academy of Agricultural Sciences, Wuhan 430064, Hubei, China
- Hubei Key Laboratory of Nutritional Quality and Safety of Agro-products, Wuhan 430064, Hubei, China
- Ministry of Agriculture and Rural Affairs Laboratory of Quality and Safe Risk Assessment for Agro-products (Wuhan), Wuhan 430064, Hubei, China
| | - Yu Shang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Youxiang Zhou
- Institute of Quality Standard and Testing Technology for Agro-Products, Hubei Academy of Agricultural Sciences, Wuhan 430064, Hubei, China
- Hubei Key Laboratory of Nutritional Quality and Safety of Agro-products, Wuhan 430064, Hubei, China
- Ministry of Agriculture and Rural Affairs Laboratory of Quality and Safe Risk Assessment for Agro-products (Wuhan), Wuhan 430064, Hubei, China
| | - Qingping Luo
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
- Hubei Hongshan Laboratory, Wuhan 430064, China
| | - Guoyuan Wen
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Zhenyu Cheng
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
4
|
Li S, Zhou Y, Yan Y, Qin Y, Weng Q, Sun L. Structure-Based Virtual Screening, ADMET Properties Prediction and Molecular Dynamics Studies Reveal Potential Inhibitors of Mycoplasma pneumoniae HPrK/P. Life (Basel) 2024; 14:657. [PMID: 38929642 PMCID: PMC11204831 DOI: 10.3390/life14060657] [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: 04/26/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Mycoplasma pneumoniae pneumonia (MPP) is a frequent cause of community-acquired pneumonia (CAP) in children. The incidence of childhood pneumonia caused by M. pneumoniae infection has been rapidly increasing worldwide. M. pneumoniae is naturally resistant to beta-lactam antibiotics due to its lack of a cell wall. Macrolides and related antibiotics are considered the optimal drugs for treating M. pneumoniae infection. However, clinical resistance to macrolides has become a global concern in recent years. Therefore, it is imperative to urgently identify new targets and develop new anti-M. pneumoniae drugs to treat MMP. Previous studies have shown that deficiencies in HPrK/P kinase or phosphorylase activity can seriously affect carbon metabolism, growth, morphology, and other cellular functions of M. pneumoniae. To identify potential drug development targets against M. pneumoniae, this study analyzed the sequence homology and 3D structure alignment of M. pneumoniae HPrK/P. Through sequence and structure analysis, we found that HPrK/P lacks homologous proteins in the human, while its functional motifs are highly conserved in bacteria. This renders it a promising candidate for drug development. Structure-based virtual screening was then used to discover potential inhibitors among 2614 FDA-approved drugs and 948 bioactive small molecules for M. pneumoniae HPrK/P. Finally, we identified three candidate drugs (Folic acid, Protokylol and Gluconolactone) as potential HPrK/P inhibitors through molecular docking, molecular dynamics (MDs) simulations, and ADMET predictions. These drugs offer new strategies for the treatment of MPP.
Collapse
Affiliation(s)
- Shen Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518107, China; (S.L.); (Y.Z.); (Y.Y.); (Y.Q.); (Q.W.)
| | - Ying Zhou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518107, China; (S.L.); (Y.Z.); (Y.Y.); (Y.Q.); (Q.W.)
| | - Yujuan Yan
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518107, China; (S.L.); (Y.Z.); (Y.Y.); (Y.Q.); (Q.W.)
| | - Yinying Qin
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518107, China; (S.L.); (Y.Z.); (Y.Y.); (Y.Q.); (Q.W.)
| | - Qilu Weng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518107, China; (S.L.); (Y.Z.); (Y.Y.); (Y.Q.); (Q.W.)
| | - Litao Sun
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518107, China; (S.L.); (Y.Z.); (Y.Y.); (Y.Q.); (Q.W.)
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen 518107, China
| |
Collapse
|
5
|
de Souza V, Polaquini CR, de Moraes GR, Oliveira Braga AR, da Silva PV, da Silva DR, Ribeiro Lima FR, Regasini LO, de Cássia Orlandi Sardi J. Diacetylcurcumin: a novel strategy against Enterococcus faecalis biofilm in root canal disinfection. Future Microbiol 2024; 19:647-654. [PMID: 38661711 PMCID: PMC11259074 DOI: 10.2217/fmb-2023-0235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/23/2024] [Indexed: 04/26/2024] Open
Abstract
Aim: We evaluated Diacetylcurcumin (DAC), a derivative of curcumin, for its antibacterial and antibiofilm properties against Enterococcus faecalis. Methods: Minimum inhibitory concentration (MIC) and minimum bactericidal concentration were determined, along with antibiofilm potential and toxicity in Galleria mellonella. Additionally, in silico computational analysis was performed to understand its mechanisms of action. Results & conclusion: DAC demonstrated significant antibacterial effects, with MIC and MBC values of 15.6 and 31.25 μg/ml, respectively, and reduced biofilm formation. A synergistic effect, reducing biofilm by 77%, was observed when combined with calcium hydroxide. G. mellonella toxicity tests confirmed DAC's safety at tested concentrations, suggesting its potential for use in root canal disinfection products.
Collapse
Affiliation(s)
- Vanessa de Souza
- Program on Integrated Dental Sciences, Cuiabá University, Cuiabá, Brazil
| | - Carlos Roberto Polaquini
- Department of Chemistry & Environmental Sciences, Júlio de Mesquita Filho University, São Jose do Rio Preto, Brazil
| | - Graciele Ribeiro de Moraes
- Department of Chemistry & Environmental Sciences, Júlio de Mesquita Filho University, São Jose do Rio Preto, Brazil
| | | | | | | | | | - Luís Octávio Regasini
- Department of Chemistry & Environmental Sciences, Júlio de Mesquita Filho University, São Jose do Rio Preto, Brazil
| | - Janaina de Cássia Orlandi Sardi
- Program on Integrated Dental Sciences, Cuiabá University, Cuiabá, Brazil
- Dental Research Division, Guarulhos University, Guarulhos, SP, Brazil
| |
Collapse
|
6
|
Enany S, Tartor YH, Kishk RM, Gadallah AM, Ahmed E, Magdeldin S. Proteomics and metabolomics analyses of Streptococcus agalactiae isolates from human and animal sources. Sci Rep 2023; 13:20980. [PMID: 38017083 PMCID: PMC10684508 DOI: 10.1038/s41598-023-47976-y] [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: 05/21/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023] Open
Abstract
Streptococcus agalactiae (S. agalactiae), group B Streptococcus (GBS), a major cause of infection in a wide variety of diseases, have been compared in different human and animal sources. We aimed to compare the bacterial proteome and metabolome profiles of human and animal S. agalactiae strains to delineate biological interactions relevant to infection. With the innovative advancement in mass spectrometry, a comparative result between both strains provided a solid impression of different responses to the host. For instance, stress-related proteins (Asp23/Gls24 family envelope stress response protein and heat shock protein 70), which play a role in the survival of GBS under extreme environmental conditions or during treatment, are highly expressed in human and animal strains. One human strain contains ꞵ-lactamase (serine hydrolase) and biofilm regulatory protein (lytR), which are important virulence regulators and potential targets for the design of novel antimicrobials. Another human strain contains the aminoglycosides-resistance bifunctional AAC/APH (A0A0U2QMQ5) protein, which confers resistance to almost all clinically used aminoglycosides. Fifteen different metabolites were annotated between the two groups. L-aspartic acid, ureidopropionic acid, adenosine monophosphate, L-tryptophan, and guanosine monophosphate were annotated at higher levels in human strains. Butyric acid, fumaric acid, isoleucine, leucine, and hippuric acid have been found in both human and animal strains. Certain metabolites were uniquely expressed in animal strains, with fold changes greater than 2. For example, putrescine modulates biofilm formation. Overall, this study provides biological insights into the substantial possible bacterial response reflected in its macromolecular production, either at the proteomic or metabolomic level.
Collapse
Affiliation(s)
- Shymaa Enany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
- Biomedical Research Department, Armed Force College of Medicine, Cairo, Egypt.
| | - Yasmine H Tartor
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Rania M Kishk
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Ahmed M Gadallah
- Department of Obstetrics and Gynecology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Eman Ahmed
- Proteomics and Metabolomics Unit, Department of Basic Research, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Sameh Magdeldin
- Proteomics and Metabolomics Unit, Department of Basic Research, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
7
|
Leasure CS, Grunenwald CM, Choby JE, Sauer JD, Skaar EP. Maintenance of heme homeostasis in Staphylococcus aureus through post-translational regulation of glutamyl-tRNA reductase. J Bacteriol 2023; 205:e0017123. [PMID: 37655914 PMCID: PMC10521356 DOI: 10.1128/jb.00171-23] [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: 06/01/2023] [Accepted: 06/30/2023] [Indexed: 09/02/2023] Open
Abstract
Staphylococcus aureus is an important human pathogen responsible for a variety of infections including skin and soft tissue infections, endocarditis, and sepsis. The combination of increasing antibiotic resistance in this pathogen and the lack of an efficacious vaccine underscores the importance of understanding how S. aureus maintains metabolic homeostasis in a variety of environments, particularly during infection. Within the host, S. aureus must regulate cellular levels of the cofactor heme to support enzymatic activities without encountering heme toxicity. Glutamyl tRNA reductase (GtrR), the enzyme catalyzing the first committed step in heme synthesis, is an important regulatory node of heme synthesis in Bacteria, Archaea, and Plantae. In many organisms, heme status negatively regulates the abundance of GtrR, controlling flux through the heme synthesis pathway. We identified two residues within GtrR, H32 and R214, that are important for GtrR-heme binding. However, in strains expressing either GtrRH32A or GtrRR214A, heme homeostasis was not perturbed, suggesting an alternative mechanism of heme synthesis regulation occurs in S. aureus. In this regard, we report that heme synthesis is regulated through phosphorylation and dephosphorylation of GtrR by the serine/threonine kinase Stk1 and the phosphatase Stp1, respectively. Taken together, these results suggest that the mechanisms governing staphylococcal heme synthesis integrate both the availability of heme and the growth status of the cell. IMPORTANCE Staphylococcus aureus represents a significant threat to human health. Heme is an iron-containing enzymatic cofactor that can be toxic at elevated levels. During infection, S. aureus must control heme levels to replicate and survive within the hostile host environment. We identified residues within a heme biosynthetic enzyme that are critical for heme binding in vitro; however, abrogation of heme binding is not sufficient to perturb heme homeostasis within S. aureus. This marks a divergence from previously reported mechanisms of heme-dependent regulation of the highly conserved enzyme glutamyl tRNA reductase (GtrR). Additionally, we link cell growth arrest to the modulation of heme levels through the post-translational regulation of GtrR by the kinase Stk1 and the phosphatase Stp1.
Collapse
Affiliation(s)
- Catherine S. Leasure
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Caroline M. Grunenwald
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jacob E. Choby
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
8
|
Konaklieva MI, Plotkin BJ. Utilization of Existing Human Kinase Inhibitors as Scaffolds in the Development of New Antimicrobials. Antibiotics (Basel) 2023; 12:1418. [PMID: 37760715 PMCID: PMC10525673 DOI: 10.3390/antibiotics12091418] [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: 08/22/2023] [Revised: 09/03/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
The prevalence and continuing expansion of drug resistance, both in clinical and community settings represents a major challenge for current antimicrobial therapy. The different approaches for addressing this challenge include (1) identification of novel antibacterials by repurposing of existing drugs originally that historically target host proteins; and (2) effect target switching through modification of existing antimicrobials. The focus of this manuscript is on these drug discovery strategies, with utility for development of new antimicrobials with different modes of action.
Collapse
Affiliation(s)
| | - Balbina J. Plotkin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA;
| |
Collapse
|
9
|
Mali SN, Anand A, Zaki MEA, Al-Hussain SA, Jawarkar RD, Pandey A, Kuznetsov A. Theoretical and Anti- Klebsiella pneumoniae Evaluations of Substituted 2,7-dimethylimidazo[1,2-a]pyridine-3-carboxamide and Imidazopyridine Hydrazide Derivatives. Molecules 2023; 28:molecules28062801. [PMID: 36985773 PMCID: PMC10051578 DOI: 10.3390/molecules28062801] [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: 02/23/2023] [Revised: 03/08/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
A series of multistep synthesis protocols was adopted to synthesize substituted imidazopyridines (IMPs) (SM-IMP-01 to SM-IMP-13, and DA-01-05). All substituted IMPs were then characterized using standard spectroscopic techniques such as 1H-NMR, 13C-NMR, elemental analyses, and mass spectrometry. Our both in vitro qualitative and quantitative results for antibacterial analysis, against Klebsiella pneumoniae ATCC 4352 and Bacillus subtilis ATCC 6051 suggested that all compounds essentially exhibited activity against selected strains of bacteria. Our DFT analyses suggested that the compounds of the SM-IMP-01-SM-IMP-13 series have HOMO/LUMO gaps within 4.43-4.69 eV, whereas the compounds of the DA-01-DA-05 series have smaller values of the HOMO/LUMO gaps, 3.24-4.17 eV. The lowest value of the global hardness and the highest value of the global softness, 2.215 and 0.226 eV, respectively, characterize the compound SM-IMP-02; thus, it is the most reactive compound in the imidazopyridine carboxamide series (except hydrazide series). This compound also depicted lesser MIC values against Klebsiella pneumoniae ATCC 4352 and Bacillus subtilis ATCC 6051 as 4.8 µg/mL, each. In terms of another series, hydrazide DA-05 depicted strong antimicrobial actions (MIC: 4.8 µg/mL against both bacterial strains) and also had the lowest energy gap (3.24 eV), higher softness (0.309 eV), and lesser hardness (1.62 eV). Overall, when we compare qualitative and quantitative antimicrobial results, it is been very clear that compounds with dibromo substitutions on imidazopyridine (IMP) rings would act as better antimicrobial agents than those with -H at the eighth position on the IMP ring. Furthermore, substituents of higher electronegativities would tend to enhance the biological activities of dibromo-IMP compounds. DFT properties were also well comparable to this trend and overall, we can say that the electronic behavior of compounds under investigation has key roles in their bioactivities.
Collapse
Affiliation(s)
- Suraj N Mali
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi 835215, India
| | - Amit Anand
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi 835215, India
| | - Magdi E A Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Sami A Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Rahul D Jawarkar
- Department of Medicinal Chemistry and Drug Discovery, Dr. Rajendra Gode Institute of Pharmacy, University Mardi Road, Amravati 444603, India
| | - Anima Pandey
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi 835215, India
| | - Aleksey Kuznetsov
- Department of Chemistry, Universidad Técnica Federico Santa Maria, Santiago 7660251, Chile
| |
Collapse
|
10
|
Li H, Li T, Hu Q, Yao Z, Li L, Huang Q, Zhou R. Inhibitors targeting the autophosphorylation of serine/threonine kinase of Streptococcus suis show potent antimicrobial activity. Front Microbiol 2022; 13:990091. [PMID: 36118193 PMCID: PMC9478340 DOI: 10.3389/fmicb.2022.990091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/09/2022] [Indexed: 11/19/2022] Open
Abstract
Antimicrobial resistance (AMR) is a global concern threatening public health. Developing novel antibiotics is one of the effective strategies to tackle AMR. Serine/threonine kinases (STKs) have been recently shown to play critical roles in the physiology and pathogenesis of several important bacterial pathogens which are regarded as a promising antimicrobial drug target. We previously reported the roles of STK in the regulation of bacterial cell division, metabolism, and pathogenesis in Streptococcus suis, an important zoonotic bacterial pathogen. In this study, we firstly identified the Thr167 and Ser175 residues in the activation loop of S. suis STK (ssSTK) as the kinase autophosphorylation sites. Phenotyping results demonstrated that the autophosphorylation deficient strain resembled the stk deletion strain showing essentiality for bacterial growth in minimal medium, abnormal morphology, and decreased virulence when compared with the wild-type S. suis SC19 strain. Based on these findings, we established an ssSTK inhibitor screening approach by measuring the growth of S. suis in a minimal medium and testing the autophosphorylation inhibition by measuring the consumption of ATP in an enzymatic reaction by ssSTK. A series of inhibitors against ssSTK are identified from a commercial kinase inhibitors library, including Staurosporine, K252a, AT9283, and APY29. These inhibitors showed antimicrobial activity in vitro. Moreover, by using Galleria mellonella larvae infection assay, compound APY29 displayed in vivo efficacy against S. suis infection. Additionally, it was predicted by molecular docking that these inhibitors could interact with ssSTK. Collectively, our data illustrated the essential roles of ssSTK autophosphorylation in the physiology and pathogenicity of S. suis and consider these inhibitors as promising antimicrobial lead compounds.
Collapse
Affiliation(s)
- Haotian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tingting Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qiao Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhiming Yao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lu Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China
- International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
| | - Qi Huang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China
- International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
- *Correspondence: Qi Huang,
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China
- International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
- The HZAU-HVSEN Institute, Wuhan, China
- Rui Zhou,
| |
Collapse
|
11
|
Kumar S, Bhadane R, Shandilya S, Salo-Ahen OMH, Kapila S. Identification of HPr kinase/phosphorylase inhibitors: novel antimicrobials against resistant Enterococcus faecalis. J Comput Aided Mol Des 2022; 36:507-520. [PMID: 35809194 PMCID: PMC9399212 DOI: 10.1007/s10822-022-00461-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 06/14/2022] [Indexed: 12/02/2022]
Abstract
Enterococcus faecalis, a gram-positive bacterium, is among the most common nosocomial pathogens due to its limited susceptibility to antibiotics and its reservoir of the genes coding for virulence factors. Bacterial enzymes such as kinases and phosphorylases play important roles in diverse functions of a bacterial cell and, thus, are potential antibacterial drug targets. In Gram-positive bacteria, HPr Kinase/Phosphorylase (HPrK/P), a bifunctional enzyme is involved in the regulation of carbon catabolite repression by phosphorylating/dephosphorylating the histidine-containing phosphocarrier protein (HPr) at Ser46 residue. Deficiencies in HPrK/P function leads to severe defects in bacterial growth. This study aimed at identifying novel inhibitors of E. faecalis HPrK/P from a commercial compound library using structure-based virtual screening. The hit molecules were purchased and their effect on enzyme activity and growth of resistant E. faecalis was evaluated in vitro. Furthermore, docking and molecular dynamics simulations were performed to study the interactions of the hit compounds with HPrK/P. Among the identified hit molecules, two compounds inhibited the phosphorylation of HPr as well as significantly reduced the growth of resistant E. faecalis in vitro. These identified potential HPrK/P inhibitors open new research avenues towards the development of novel antimicrobials against resistant Gram-positive bacteria.
Collapse
Affiliation(s)
- Sandeep Kumar
- Animal Biochemistry Division, National Dairy Research Institute, Karnal, Haryana, India
| | - Rajendra Bhadane
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, 20520, Turku, Finland
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Pharmacy, Åbo Akademi University, 20520, Turku, Finland
| | - Shruti Shandilya
- Department of Applied Physics, School of Science, Aalto University, Espoo, Finland
| | - Outi M H Salo-Ahen
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, 20520, Turku, Finland.
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Pharmacy, Åbo Akademi University, 20520, Turku, Finland.
| | - Suman Kapila
- Animal Biochemistry Division, National Dairy Research Institute, Karnal, Haryana, India.
| |
Collapse
|
12
|
Berndsen R, Cunningham T, Kaelin L, Callender M, Boldog WD, Viering B, King A, Labban N, Pollock JA, Miller HB, Blackledge MS. Identification and Evaluation of Brominated Carbazoles as a Novel Antibiotic Adjuvant Scaffold in MRSA. ACS Med Chem Lett 2022; 13:483-491. [PMID: 35295086 PMCID: PMC8919279 DOI: 10.1021/acsmedchemlett.1c00680] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/02/2022] [Indexed: 12/23/2022] Open
Abstract
![]()
Antibiotic-resistant
infections are a pressing global concern,
causing millions of deaths each year. Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of nosocomial
infections in healthcare settings and is increasingly responsible
for community-acquired infections that are often more difficult to
treat. Antibiotic adjuvants are small molecules that potentiate antibiotics
through nontoxic mechanisms and show excellent promise as novel therapeutics.
Screening of low-molecular-weight compounds was employed to identify
novel antibiotic adjuvant scaffolds for further elaboration. Brominated
carbazoles emerged from this screening as lead compounds for further
evaluation. Lead carbazoles were able to potentiate several β-lactam
antibiotics in three medically relevant strains of MRSA. Gene expression
studies determined that these carbazoles were dampening the transcription
of key genes that modulate β-lactam resistance in MRSA. The
lead brominated carbazoles represent novel scaffolds for elaboration
as antibiotic adjuvants.
Collapse
Affiliation(s)
- Rachel Berndsen
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| | - Taylor Cunningham
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| | - Lauren Kaelin
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| | - Makayla Callender
- Department of Chemistry, University of Richmond, Richmond, Virginia 23173, United States
| | - W. Dexter Boldog
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| | - Brianna Viering
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| | - Ashley King
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| | - Najwa Labban
- Department of Chemistry, University of Richmond, Richmond, Virginia 23173, United States
| | - Julie A. Pollock
- Department of Chemistry, University of Richmond, Richmond, Virginia 23173, United States
| | - Heather B. Miller
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| | - Meghan S. Blackledge
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| |
Collapse
|
13
|
Ali IO, Salama TM, A. Gawad A, El‐Henawy AA, Ghazy M, Bakr MF. Silver nanoparticles @ titanate nanotubes composite: Synthesis, characterization, applications and docking. INORG CHEM COMMUN 2022. [DOI: 10.1016/j.inoche.2021.109187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
14
|
Hamad M, Al-Marzooq F, Srinivasulu V, Omar HA, Sulaiman A, Zaher DM, Orive G, Al-Tel TH. Antibacterial Activity of Small Molecules Which Eradicate Methicillin-Resistant Staphylococcus aureus Persisters. Front Microbiol 2022; 13:823394. [PMID: 35178043 PMCID: PMC8846302 DOI: 10.3389/fmicb.2022.823394] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
The serious challenge posed by multidrug-resistant bacterial infections with concomitant treatment failure and high mortality rates presents an urgent threat to the global health. We herein report the discovery of a new class of potent antimicrobial compounds that are highly effective against Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA). The compounds were efficiently synthesized in one-pot employing a cascade of Groebke-Blackburn-Bienaymé and aza-Michael addition reactions. Phenotypic screening of the pilot library against various bacterial species including methicillin-sensitive and MRSA strains, has identified potent chemotypes with minimal inhibitory concentrations (MIC) of 3.125-6.25 μg/ml. The most potent compounds were fast-acting at eradicating exponentially growing MRSA, with killing achieved after 30 min of exposure to the compounds. They were also able to kill MRSA persister cells which are tolerant to most available medications. Microscopic analysis using fluorescence microscope and atomic force microscope indicate that these compounds lead to disruption of bacterial cell envelopes. Most notably, bacterial resistance toward these compounds was not observed after 20 serial passages in stark contrast to the significant resistance developed rapidly upon exposure to a clinically relevant antibiotic. Furthermore, the compounds did not induce significant hemolysis to human red blood cells. In vivo safety studies revealed a high safety profile of these motifs. These small molecules hold a promise for further studies and development as new antibacterial agents against MRSA infections.
Collapse
Affiliation(s)
- Mohamad Hamad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Farah Al-Marzooq
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Vunnam Srinivasulu
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Hany A Omar
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ashna Sulaiman
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Dana M Zaher
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Taleb H Al-Tel
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
15
|
King A, Blackledge MS. Evaluation of small molecule kinase inhibitors as novel antimicrobial and antibiofilm agents. Chem Biol Drug Des 2021; 98:1038-1064. [PMID: 34581492 PMCID: PMC8616828 DOI: 10.1111/cbdd.13962] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/24/2021] [Accepted: 09/15/2021] [Indexed: 12/25/2022]
Abstract
Antibiotic resistance is a global and pressing concern. Our current therapeutic arsenal is increasingly limited as bacteria are developing resistance at a rate that far outpaces our ability to create new treatments. Novel approaches to treating and curing bacterial infections are urgently needed. Bacterial kinases have been increasingly explored as novel drug targets and are poised for development into novel therapeutic agents to combat bacterial infections. This review describes several general classes of bacterial kinases that play important roles in bacterial growth, antibiotic resistance, and biofilm formation. General features of these kinase classes are discussed and areas of particular interest for the development of inhibitors will be highlighted. Small molecule kinase inhibitors are described and organized by phenotypic effect, spotlighting particularly interesting inhibitors with novel functions and potential therapeutic benefit. Finally, we provide our perspective on the future of bacterial kinase inhibition as a viable strategy to combat bacterial infections and overcome the pressures of increasing antibiotic resistance.
Collapse
Affiliation(s)
- Ashley King
- Department of Chemistry, High Point University, One University Parkway, High Point, NC 27268
| | - Meghan S. Blackledge
- Department of Chemistry, High Point University, One University Parkway, High Point, NC 27268
| |
Collapse
|
16
|
Kelliher JL, Grunenwald CM, Abrahams RR, Daanen ME, Lew CI, Rose WE, Sauer JD. PASTA kinase-dependent control of peptidoglycan synthesis via ReoM is required for cell wall stress responses, cytosolic survival, and virulence in Listeria monocytogenes. PLoS Pathog 2021; 17:e1009881. [PMID: 34624065 PMCID: PMC8528326 DOI: 10.1371/journal.ppat.1009881] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/20/2021] [Accepted: 09/27/2021] [Indexed: 02/01/2023] Open
Abstract
Pathogenic bacteria rely on protein phosphorylation to adapt quickly to stress, including that imposed by the host during infection. Penicillin-binding protein and serine/threonine-associated (PASTA) kinases are signal transduction systems that sense cell wall integrity and modulate multiple facets of bacterial physiology in response to cell envelope stress. The PASTA kinase in the cytosolic pathogen Listeria monocytogenes, PrkA, is required for cell wall stress responses, cytosolic survival, and virulence, yet its substrates and downstream signaling pathways remain incompletely defined. We combined orthogonal phosphoproteomic and genetic analyses in the presence of a β-lactam antibiotic to define PrkA phosphotargets and pathways modulated by PrkA. These analyses synergistically highlighted ReoM, which was recently identified as a PrkA target that influences peptidoglycan (PG) synthesis, as an important phosphosubstrate during cell wall stress. We find that deletion of reoM restores cell wall stress sensitivities and cytosolic survival defects of a ΔprkA mutant to nearly wild-type levels. While a ΔprkA mutant is defective for PG synthesis during cell wall stress, a double ΔreoM ΔprkA mutant synthesizes PG at rates similar to wild type. In a mouse model of systemic listeriosis, deletion of reoM in a ΔprkA background almost fully restored virulence to wild-type levels. However, loss of reoM alone also resulted in attenuated virulence, suggesting ReoM is critical at some points during pathogenesis. Finally, we demonstrate that the PASTA kinase/ReoM cell wall stress response pathway is conserved in a related pathogen, methicillin-resistant Staphylococcus aureus. Taken together, our phosphoproteomic analysis provides a comprehensive overview of the PASTA kinase targets of an important model pathogen and suggests that a critical role of PrkA in vivo is modulating PG synthesis through regulation of ReoM to facilitate cytosolic survival and virulence. Many antibiotics target bacterial cell wall biosynthesis, justifying continued study of this process and the ways bacteria respond to cell wall insults during infection. Penicillin-binding protein and serine/threonine-associated (PASTA) kinases are master regulators of cell wall stress responses in bacteria and are conserved in several major pathogens, including Listeria monocytogenes, Staphylococcus aureus, and Mycobacterium tuberculosis. We previously showed that the PASTA kinase in L. monocytogenes, PrkA, is essential for the response to cell wall stress and for virulence. In this work, we combined proteomic and genetic approaches to identify PrkA substrates in L. monocytogenes. We show that regulation of one candidate from both screens, ReoM, increases synthesis of the cell wall component peptidoglycan and that this regulation is required for pathogenesis. We also demonstrate that the PASTA kinase-ReoM pathway regulates cell wall stress responses in another significant pathogen, methicillin-resistant S. aureus. Additionally, we uncover a PrkA-independent role for ReoM in vivo in L. monocytogenes, suggesting a need for nuanced modulation of peptidoglycan synthesis during infection. Cumulatively, this study provides new insight into how bacterial pathogens control cell wall synthesis during infection.
Collapse
Affiliation(s)
- Jessica L. Kelliher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Caroline M. Grunenwald
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Rhiannon R. Abrahams
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - McKenzie E. Daanen
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Cassandra I. Lew
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Warren E. Rose
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
17
|
RAHAL ANU, KUMAR AMIT. Strategies to combat antimicrobial resistance in Indian scenario. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2021. [DOI: 10.56093/ijans.v91i2.113812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Antimicrobial resistance (AMR) is one of the major public health crisis recognised globally. Microbial infections cause significant productivity losses in animals and humans. In livestock, these microbial infections reduce the growth rates and fertility, diminish production of meat and milk, and occasionally lead to mortality, and are therefore, a major concern for animal welfare. In the dearth of alternative prophylactic measures, antibiotics remain the principal tool for their management. Once an antibiotic is used rampantly, resistance against it is inevidently seen in the microbe population and the hunt for a new drug grows. Discovery and development of a new antimicrobial drug is a time taking and expensive procedure with limited assurance of success. As a result, the past few decades have witnessed only a very few new classes of antibiotics. If the AMR can be restricted or reverted, the success rate of antimicrobial therapy can be boosted and many public health issues be avoided. All these ask for a comprehensive plan to prevent or reduce the antimicrobial resistance and economic losses to the animal husbandry sector. The present review provides an overview of AMR in India, mechanism of its occurrence and the possible roadmap to combat the emerging threat of AMR in Indian scenario.
Collapse
|
18
|
Fisher JF, Mobashery S. β-Lactams against the Fortress of the Gram-Positive Staphylococcus aureus Bacterium. Chem Rev 2021; 121:3412-3463. [PMID: 33373523 PMCID: PMC8653850 DOI: 10.1021/acs.chemrev.0c01010] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The biological diversity of the unicellular bacteria-whether assessed by shape, food, metabolism, or ecological niche-surely rivals (if not exceeds) that of the multicellular eukaryotes. The relationship between bacteria whose ecological niche is the eukaryote, and the eukaryote, is often symbiosis or stasis. Some bacteria, however, seek advantage in this relationship. One of the most successful-to the disadvantage of the eukaryote-is the small (less than 1 μm diameter) and nearly spherical Staphylococcus aureus bacterium. For decades, successful clinical control of its infection has been accomplished using β-lactam antibiotics such as the penicillins and the cephalosporins. Over these same decades S. aureus has perfected resistance mechanisms against these antibiotics, which are then countered by new generations of β-lactam structure. This review addresses the current breadth of biochemical and microbiological efforts to preserve the future of the β-lactam antibiotics through a better understanding of how S. aureus protects the enzyme targets of the β-lactams, the penicillin-binding proteins. The penicillin-binding proteins are essential enzyme catalysts for the biosynthesis of the cell wall, and understanding how this cell wall is integrated into the protective cell envelope of the bacterium may identify new antibacterials and new adjuvants that preserve the efficacy of the β-lactams.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| |
Collapse
|
19
|
Are antibacterial effects of non-antibiotic drugs random or purposeful because of a common evolutionary origin of bacterial and mammalian targets? Infection 2020; 49:569-589. [PMID: 33325009 PMCID: PMC7737717 DOI: 10.1007/s15010-020-01547-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/28/2020] [Indexed: 01/09/2023]
Abstract
Purpose Advances in structural biology, genetics, bioinformatics, etc. resulted in the availability of an enormous pool of information enabling the analysis of the ancestry of pro- and eukaryotic genes and proteins. Methods This review summarizes findings of structural and/or functional homologies of pro- and eukaryotic enzymes catalysing analogous biological reactions because of their highly conserved active centres so that non-antibiotics interacted with bacterial targets. Results Protease inhibitors such as staurosporine or camostat inhibited bacterial serine/threonine or serine/tyrosine protein kinases, serine/threonine phosphatases, and serine/threonine kinases, to which penicillin-binding-proteins are linked, so that these drugs synergized with β-lactams, reverted aminoglycoside-resistance and attenuated bacterial virulence. Calcium antagonists such as nitrendipine or verapamil blocked not only prokaryotic ion channels but interacted with negatively charged bacterial cell membranes thus disrupting membrane energetics and inducing membrane stress response resulting in inhibition of P-glycoprotein such as bacterial pumps thus improving anti-mycobacterial activities of rifampicin, tetracycline, fluoroquinolones, bedaquilin and imipenem-activity against Acinetobacter spp. Ciclosporine and tacrolimus attenuated bacterial virulence. ACE-inhibitors like captopril interacted with metallo-β-lactamases thus reverting carbapenem-resistance; prokaryotic carbonic anhydrases were inhibited as well resulting in growth impairment. In general, non-antibiotics exerted weak antibacterial activities on their own but synergized with antibiotics, and/or reverted resistance and/or attenuated virulence. Conclusions Data summarized in this review support the theory that prokaryotic proteins represent targets for non-antibiotics because of a common evolutionary origin of bacterial- and mammalian targets resulting in highly conserved active centres of both, pro- and eukaryotic proteins with which the non-antibiotics interact and exert antibacterial actions.
Collapse
|
20
|
Greco C, Catania R, Balacco DL, Taresco V, Musumeci F, Alexander C, Huett A, Schenone S. Synthesis and Antibacterial Evaluation of New Pyrazolo[3,4- d]pyrimidines Kinase Inhibitors. Molecules 2020; 25:molecules25225354. [PMID: 33207806 PMCID: PMC7696985 DOI: 10.3390/molecules25225354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
Pyrazolo[3,4-d]pyrimidines represent an important class of heterocyclic compounds well-known for their anticancer activity exerted by the inhibition of eukaryotic protein kinases. Recently, pyrazolo[3,4-d]pyrimidines have become increasingly attractive for their potential antimicrobial properties. Here, we explored the activity of a library of in-house pyrazolo[3,4-d]pyrimidines, targeting human protein kinases, against Staphylococcus aureus and Escherichia coli and their interaction with ampicillin and kanamycin, representing important classes of clinically used antibiotics. Our results represent a first step towards the potential application of dual active pyrazolo[3,4-d]pyrimidine kinase inhibitors in the prevention and treatment of bacterial infections in cancer patients.
Collapse
Affiliation(s)
- Chiara Greco
- Dipartimento di Farmacia, Università di Genova, Viale Benedetto XV 3, 16132 Genova, Italy; (C.G.); (F.M.)
| | - Rosa Catania
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
| | - Dario Leonardo Balacco
- School of Dentistry, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B5 7EG, UK;
| | - Vincenzo Taresco
- School of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, UK;
| | - Francesca Musumeci
- Dipartimento di Farmacia, Università di Genova, Viale Benedetto XV 3, 16132 Genova, Italy; (C.G.); (F.M.)
| | - Cameron Alexander
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK;
| | - Alan Huett
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
- Correspondence: (A.H.); (S.S.)
| | - Silvia Schenone
- Dipartimento di Farmacia, Università di Genova, Viale Benedetto XV 3, 16132 Genova, Italy; (C.G.); (F.M.)
- Correspondence: (A.H.); (S.S.)
| |
Collapse
|
21
|
Djorić D, Minton NE, Kristich CJ. The enterococcal PASTA kinase: A sentinel for cell envelope stress. Mol Oral Microbiol 2020; 36:132-144. [PMID: 32945615 DOI: 10.1111/omi.12313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/05/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022]
Abstract
Enterococci are Gram-positive, opportunistic pathogens that reside throughout the gastrointestinal tracts of most terrestrial organisms. Enterococci are resistant to many antibiotics, which makes enterococcal infections difficult to treat. Enterococci are also particularly hardy bacteria that can tolerate a variety of environmental stressors. Understanding how enterococci sense and respond to the extracellular environment to enact adaptive biological responses may identify new targets that can be exploited for development of treatments for enterococcal infections. Bacterial eukaryotic-like serine/threonine kinases (eSTKs) and cognate phosphatases (STPs) are important signaling systems that mediate biological responses to extracellular stimuli. Some bacterial eSTKs are transmembrane proteins that contain a series of extracellular repeats of the penicillin-binding and Ser/Thr kinase-associated (PASTA) domain, leading to their designation as "PASTA kinases." Enterococcal genomes encode a single PASTA kinase and its cognate phosphatase. Investigations of the enterococcal PASTA kinase revealed its importance in resistance to antibiotics and other cell wall stresses, in enterococcal colonization of the mammalian gut, clues about its mechanism of signal transduction, and its integration with other enterococcal signal transduction systems. In this review, we describe the current state of knowledge of PASTA kinase signaling in enterococci and describe important gaps that still need to be addressed to provide a better understanding of this important signaling system.
Collapse
Affiliation(s)
- Dušanka Djorić
- Department of Microbiology and Immunology, Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nicole E Minton
- Department of Microbiology and Immunology, Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christopher J Kristich
- Department of Microbiology and Immunology, Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
22
|
Regulation of virulence and antibiotic resistance in Gram-positive microbes in response to cell wall-active antibiotics. Curr Opin Infect Dis 2020; 32:217-222. [PMID: 31021953 DOI: 10.1097/qco.0000000000000542] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Antibiotic stress can evoke considerable genotypic and phenotypic changes in Gram-positive bacteria. Here, we review recent studies describing altered virulence expression in response to cell wall-acting antibiotics and discuss mechanisms that coordinate regulation of the antibiotic response. RECENT FINDINGS Pleiotropic effects induced by antibiotic exposure include alterations to bacterial metabolism, cell wall structure and antibiotic resistance. In addition, subinhibitory concentrations of cell wall-active (CWA) antibiotics have increasingly been shown to induce the production of exotoxins and biofilm formation that may influence virulence. Remarkably, phenotypes associated with comparable antibiotic stresses can vary considerably, emphasizing the need to better understand the response to CWA antibiotics. Recent studies support both direct antibiotic recognition and recognition of antibiotic-induced stress to the bacterial cell wall. Specifically, bacterial two-component systems, penicillin-binding protein and serine/threonine kinase-associated kinases and conserved oxidative-stress sensors each contribute to modulating the antibiotic stress response. SUMMARY Bacterial sensory systems and global regulators coordinate signaling in response to CWA antibiotics. Regulation of the antibiotic response is complex and involves integration of signals from multiple response pathways. A better definition of the antibiotic stress response among Gram-positive pathogens may yield novel therapeutic targets to counter antibiotic resistance and virulence factor expression.
Collapse
|
23
|
Bonne Køhler J, Jers C, Senissar M, Shi L, Derouiche A, Mijakovic I. Importance of protein Ser/Thr/Tyr phosphorylation for bacterial pathogenesis. FEBS Lett 2020; 594:2339-2369. [PMID: 32337704 DOI: 10.1002/1873-3468.13797] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022]
Abstract
Protein phosphorylation regulates a large variety of biological processes in all living cells. In pathogenic bacteria, the study of serine, threonine, and tyrosine (Ser/Thr/Tyr) phosphorylation has shed light on the course of infectious diseases, from adherence to host cells to pathogen virulence, replication, and persistence. Mass spectrometry (MS)-based phosphoproteomics has provided global maps of Ser/Thr/Tyr phosphosites in bacterial pathogens. Despite recent developments, a quantitative and dynamic view of phosphorylation events that occur during bacterial pathogenesis is currently lacking. Temporal, spatial, and subpopulation resolution of phosphorylation data is required to identify key regulatory nodes underlying bacterial pathogenesis. Herein, we discuss how technological improvements in sample handling, MS instrumentation, data processing, and machine learning should improve bacterial phosphoproteomic datasets and the information extracted from them. Such information is expected to significantly extend the current knowledge of Ser/Thr/Tyr phosphorylation in pathogenic bacteria and should ultimately contribute to the design of novel strategies to combat bacterial infections.
Collapse
Affiliation(s)
- Julie Bonne Køhler
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Carsten Jers
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Mériem Senissar
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Lei Shi
- Systems and Synthetic Biology Division, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Abderahmane Derouiche
- Systems and Synthetic Biology Division, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Ivan Mijakovic
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark.,Systems and Synthetic Biology Division, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
24
|
Wang CH, Hsieh YH, Powers ZM, Kao CY. Defeating Antibiotic-Resistant Bacteria: Exploring Alternative Therapies for a Post-Antibiotic Era. Int J Mol Sci 2020; 21:E1061. [PMID: 32033477 PMCID: PMC7037027 DOI: 10.3390/ijms21031061] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Antibiotics are one of the greatest medical advances of the 20th century, however, they are quickly becoming useless due to antibiotic resistance that has been augmented by poor antibiotic stewardship and a void in novel antibiotic discovery. Few novel classes of antibiotics have been discovered since 1960, and the pipeline of antibiotics under development is limited. We therefore are heading for a post-antibiotic era in which common infections become untreatable and once again deadly. There is thus an emergent need for both novel classes of antibiotics and novel approaches to treatment, including the repurposing of existing drugs or preclinical compounds and expanded implementation of combination therapies. In this review, we highlight to utilize alternative drug targets/therapies such as combinational therapy, anti-regulator, anti-signal transduction, anti-virulence, anti-toxin, engineered bacteriophages, and microbiome, to defeat antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Chih-Hung Wang
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung 40201, Taiwan;
| | - Zachary M. Powers
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Cheng-Yen Kao
- Institute of Microbiology and Immunology, School of Life Science, National Yang-Ming University, Taipei 11221, Taiwan
| |
Collapse
|
25
|
Srinivasulu V, Khanfar M, Omar HA, ElAwady R, Sieburth SM, Sebastian A, Zaher DM, Al-Marzooq F, Hersi F, Al-Tel TH. Sequencing [4 + 1]-Cycloaddition and Aza-Michael Addition Reactions: A Diastereoselective Cascade for the Rapid Access of Pyrido[2′,1′:2,3]/Thiazolo[2′,3′:2,3]imidazo[1,5-a]quinolone Scaffolds as Potential Antibacterial and Anticancer Motifs. J Org Chem 2019; 84:14476-14486. [DOI: 10.1021/acs.joc.9b01919] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Vunnam Srinivasulu
- Sharjah Institute for Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Monther Khanfar
- Department of Chemistry, University of Jordan, 11942 Amman, Jordan
| | - Hany A. Omar
- Sharjah Institute for Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Raafat ElAwady
- Sharjah Institute for Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Scott McN Sieburth
- Temple University, Department of Chemistry, 201 Beury Hall, Philadelphia, Pennsylvania 19122, United States
| | - Anusha Sebastian
- Sharjah Institute for Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Dana M. Zaher
- Sharjah Institute for Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Farah Al-Marzooq
- Sharjah Institute for Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Fatema Hersi
- Sharjah Institute for Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| | - Taleb H. Al-Tel
- Sharjah Institute for Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE
| |
Collapse
|
26
|
Speri E, Kim C, De Benedetti S, Qian Y, Lastochkin E, Fishovitz J, Fisher JF, Mobashery S. Cinnamonitrile Adjuvants Restore Susceptibility to β-Lactams against Methicillin-Resistant Staphylococcus aureus. ACS Med Chem Lett 2019; 10:1148-1153. [PMID: 31413798 PMCID: PMC6691485 DOI: 10.1021/acsmedchemlett.9b00169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 07/01/2019] [Indexed: 11/28/2022] Open
Abstract
β-Lactams are used routinely to treat Staphylococcus aureus infections. However, the emergence of methicillin-resistant S. aureus (MRSA) renders them clinically precarious. We describe a class of cinnamonitrile adjuvants that restore the activity of oxacillin (a penicillin member of the β-lactams) against MRSA. The lead adjuvants were tested against six important strains of MRSA, one vancomycin-intermediate S. aureus (VISA) strain, and one linezolid-resistant S. aureus strain. Five compounds out of 84 total compounds showed broad potentiation. At 8 μM (E)-3-(5-(3,4-dichlorobenzyl)-2-(trifluoromethoxy)phenyl)-2-(methylsulfonyl)acrylonitrile (26) potentiated oxacillin with a >4000-fold reduction of its MIC (from 256 to 0.06 mg·L-1). This class of adjuvants holds promise for reversal of the resistance phenotype of MRSA.
Collapse
Affiliation(s)
- Enrico Speri
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Choon Kim
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Stefania De Benedetti
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Yuanyuan Qian
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Elena Lastochkin
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Jennifer Fishovitz
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Jed F. Fisher
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Shahriar Mobashery
- Department Chemistry and
Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
27
|
Shahbaaz M, Nkaule A, Christoffels A. Designing novel possible kinase inhibitor derivatives as therapeutics against Mycobacterium tuberculosis: An in silico study. Sci Rep 2019; 9:4405. [PMID: 30867456 PMCID: PMC6416319 DOI: 10.1038/s41598-019-40621-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 02/18/2019] [Indexed: 11/30/2022] Open
Abstract
Rv2984 is one of the polyphosphate kinases present in Mycobacterium tuberculosis involved in the catalytic synthesis of inorganic polyphosphate, which plays an essential role in bacterial virulence and drug resistance. Consequently, the structure of Rv2984 was investigated and an 18 membered compound library was designed by altering the scaffolds of computationally identified inhibitors. The virtual screening of these altered inhibitors was performed against Rv2984 and the top three scoring inhibitors were selected, exhibiting the free energy of binding between 8.2–9 kcal mol−1 and inhibition constants in the range of 255–866 nM. These selected molecules showed relatively higher binding affinities against Rv2984 compared to the first line drugs Isoniazid and Rifampicin. Furthermore, the docked complexes were further analyzed in explicit water conditions using 100 ns Molecular Dynamics simulations. Through the assessment of obtained trajectories, the interactions between the protein and selected inhibitors including first line drugs were evaluated using MM/PBSA technique. The results validated the higher efficiency of the designed molecules compared to 1st line drugs with total interaction energies observed between −100 kJ mol−1 and −1000 kJ mol−1. This study will facilitate the process of drug designing against M. tuberculosis and can be used in the development of potential therapeutics against drug-resistant strains of bacteria.
Collapse
Affiliation(s)
- Mohd Shahbaaz
- South African National Bioinformatics Institute (SANBI), SA Medical Research Council Bioinformatics Unit, University of the Western Cape, Private Bag X17, Bellville, 7535, Cape Town, South Africa
| | - Anati Nkaule
- South African National Bioinformatics Institute (SANBI), SA Medical Research Council Bioinformatics Unit, University of the Western Cape, Private Bag X17, Bellville, 7535, Cape Town, South Africa
| | - Alan Christoffels
- South African National Bioinformatics Institute (SANBI), SA Medical Research Council Bioinformatics Unit, University of the Western Cape, Private Bag X17, Bellville, 7535, Cape Town, South Africa.
| |
Collapse
|
28
|
Wlodarchak N, Teachout N, Beczkiewicz J, Procknow R, Schaenzer AJ, Satyshur K, Pavelka M, Zuercher W, Drewry D, Sauer JD, Striker R. In Silico Screen and Structural Analysis Identifies Bacterial Kinase Inhibitors which Act with β-Lactams To Inhibit Mycobacterial Growth. Mol Pharm 2018; 15:5410-5426. [PMID: 30285456 PMCID: PMC6648700 DOI: 10.1021/acs.molpharmaceut.8b00905] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
New tools and concepts are needed to combat antimicrobial resistance. Actinomycetes and firmicutes share several eukaryotic-like Ser/Thr kinases (eSTK) that offer antibiotic development opportunities, including PknB, an essential mycobacterial eSTK. Despite successful development of potent biochemical PknB inhibitors by many groups, clinically useful microbiologic activity has been elusive. Additionally, PknB kinetics are not fully described, nor are structures with specific inhibitors available to inform inhibitor design. We used computational modeling with available structural information to identify human kinase inhibitors predicted to bind PknB, and we selected hits based on drug-like characteristics intended to increase the likelihood of cell entry. The computational model suggested a family of inhibitors, the imidazopyridine aminofurazans (IPAs), bind PknB with high affinity. We performed an in-depth characterization of PknB and found that these inhibitors biochemically inhibit PknB, with potency roughly following the predicted models. A novel X-ray structure confirmed that the inhibitors bound as predicted and made favorable protein contacts with the target. These inhibitors also have antimicrobial activity toward mycobacteria and nocardia. We demonstrated that the inhibitors are uniquely potentiated by β-lactams but not antibiotics traditionally used to treat mycobacteria, consistent with PknB's role in sensing cell wall stress. This is the first demonstration in the phylum actinobacteria that some β-lactam antibiotics could be more effective if paired with a PknB inhibitor. Collectively, our data show that in silico modeling can be used as a tool to discover promising drug leads, and the inhibitors we discovered can act with clinically relevant antibiotics to restore their efficacy against bacteria with limited treatment options.
Collapse
Affiliation(s)
- Nathan Wlodarchak
- Department of Medicine, University of Wisconsin-Madison, 3341 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Nathan Teachout
- Department of Medicine, University of Wisconsin-Madison, 3341 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Jeffrey Beczkiewicz
- Department of Medicine, University of Wisconsin-Madison, 3341 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Rebecca Procknow
- Department of Medicine, University of Wisconsin-Madison, 3341 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Adam J. Schaenzer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 4203 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Kenneth Satyshur
- Small Molecule Screening Facility, Carbone Cancer Center, University of Wisconsin-Madison, 1111Highland Ave., Madison, WI 53705
| | - Martin Pavelka
- School of Medicine and Dentistry, University of Rochester Medical Center, 601 Elmwood Ave., Rochester, NY 14620
| | - William Zuercher
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, SGC Center for Chemical Biology, 120 Mason Farm Rd., Chapel Hill, NC 27599
| | - David Drewry
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, SGC Center for Chemical Biology, 120 Mason Farm Rd., Chapel Hill, NC 27599
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 4203 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Rob Striker
- Department of Medicine, University of Wisconsin-Madison, 3341 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706,William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terr., Madison, WI 53705,To whom correspondence should be addressed Rob Striker, Department of Medicine, University of Wisconsin-Madison, 3301 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706, 608-263-2994,
| |
Collapse
|
29
|
Schaenzer AJ, Wlodarchak N, Drewry DH, Zuercher WJ, Rose WE, Ferrer CA, Sauer JD, Striker R. GW779439X and Its Pyrazolopyridazine Derivatives Inhibit the Serine/Threonine Kinase Stk1 and Act As Antibiotic Adjuvants against β-Lactam-Resistant Staphylococcus aureus. ACS Infect Dis 2018; 4:1508-1518. [PMID: 30059625 PMCID: PMC6779124 DOI: 10.1021/acsinfecdis.8b00136] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As antibiotic resistance rises, there is a need for strategies such as antibiotic adjuvants to conserve already-established antibiotics. A family of bacterial kinases known as the penicillin-binding-protein and serine/threonine kinase-associated (PASTA) kinases has attracted attention as targets for antibiotic adjuvants for β-lactams. Here, we report that the pyrazolopyridazine GW779439X sensitizes methicillin-resistant Staphylococcus aureus (MRSA) to various β-lactams through inhibition of the PASTA kinase Stk1. GW779439X potentiates β-lactam activity against multiple MRSA and MSSA isolates, including the sensitization of a ceftaroline-resistant isolate to ceftaroline. In silico modeling was used to guide the synthesis of GW779439X derivatives. The presence and orientation of GW779439X's methylpiperazine moiety was crucial for robust biochemical and microbiologic activity. Taken together, our data provide a proof of concept for developing the pyrazolopyridazines as selective Stk1 inhibitors which act across S. aureus isolates.
Collapse
Affiliation(s)
- Adam J. Schaenzer
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States
- Department of Medicine, University of Wisconsin–Madison, 1685 Highland Avenue, Madison, Wisconsin 53706, United States
| | - Nathan Wlodarchak
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States
- Department of Medicine, University of Wisconsin–Madison, 1685 Highland Avenue, Madison, Wisconsin 53706, United States
| | - David H. Drewry
- UNC Eshelman School of Pharmacy, SGC Center for Chemical Biology, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - William J. Zuercher
- UNC Eshelman School of Pharmacy, SGC Center for Chemical Biology, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Warren E. Rose
- Department of Medicine, University of Wisconsin–Madison, 1685 Highland Avenue, Madison, Wisconsin 53706, United States
- School of Pharmacy, University of Wisconsin–Madison, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Carla A. Ferrer
- UNC Eshelman School of Pharmacy, SGC Center for Chemical Biology, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States
| | - Rob Striker
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States
- Department of Medicine, University of Wisconsin–Madison, 1685 Highland Avenue, Madison, Wisconsin 53706, United States
- Department of Medicine, W. S. Middleton Memorial Veteran’s Hospital, 2500 Overlook Terrace, Madison, Wisconsin 53705, United States
| |
Collapse
|
30
|
Ramos PIP, Fernández Do Porto D, Lanzarotti E, Sosa EJ, Burguener G, Pardo AM, Klein CC, Sagot MF, de Vasconcelos ATR, Gales AC, Marti M, Turjanski AG, Nicolás MF. An integrative, multi-omics approach towards the prioritization of Klebsiella pneumoniae drug targets. Sci Rep 2018; 8:10755. [PMID: 30018343 PMCID: PMC6050338 DOI: 10.1038/s41598-018-28916-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 06/27/2018] [Indexed: 02/07/2023] Open
Abstract
Klebsiella pneumoniae (Kp) is a globally disseminated opportunistic pathogen that can cause life-threatening infections. It has been found as the culprit of many infection outbreaks in hospital environments, being particularly aggressive towards newborns and adults under intensive care. Many Kp strains produce extended-spectrum β-lactamases, enzymes that promote resistance against antibiotics used to fight these infections. The presence of other resistance determinants leading to multidrug-resistance also limit therapeutic options, and the use of 'last-resort' drugs, such as polymyxins, is not uncommon. The global emergence and spread of resistant strains underline the need for novel antimicrobials against Kp and related bacterial pathogens. To tackle this great challenge, we generated multiple layers of 'omics' data related to Kp and prioritized proteins that could serve as attractive targets for antimicrobial development. Genomics, transcriptomics, structuromic and metabolic information were integrated in order to prioritize candidate targets, and this data compendium is freely available as a web server. Twenty-nine proteins with desirable characteristics from a drug development perspective were shortlisted, which participate in important processes such as lipid synthesis, cofactor production, and core metabolism. Collectively, our results point towards novel targets for the control of Kp and related bacterial pathogens.
Collapse
Affiliation(s)
- Pablo Ivan Pereira Ramos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | - Darío Fernández Do Porto
- Plataforma de Bioinformática Argentina (BIA), Instituto de Cálculo, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA, Ciudad de Buenos Aires, Argentina
| | - Esteban Lanzarotti
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA, Ciudad de Buenos Aires, Argentina
| | - Ezequiel J Sosa
- Plataforma de Bioinformática Argentina (BIA), Instituto de Cálculo, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Germán Burguener
- Plataforma de Bioinformática Argentina (BIA), Instituto de Cálculo, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Agustín M Pardo
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA, Ciudad de Buenos Aires, Argentina
| | - Cecilia C Klein
- Inria Grenoble Rhône-Alpes, Grenoble, France
- Université Claude Bernard Lyon 1, Lyon, France
- Centre for Genomic Regulation (CRG), Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Marie-France Sagot
- Inria Grenoble Rhône-Alpes, Grenoble, France
- Université Claude Bernard Lyon 1, Lyon, France
| | | | - Ana Cristina Gales
- Laboratório Alerta. Division of Infectious Diseases, Department of Internal Medicine. Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marcelo Marti
- Plataforma de Bioinformática Argentina (BIA), Instituto de Cálculo, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA, Ciudad de Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA, Ciudad de Buenos Aires, Argentina
| | - Adrián G Turjanski
- Plataforma de Bioinformática Argentina (BIA), Instituto de Cálculo, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA, Ciudad de Buenos Aires, Argentina.
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Ciudad Universitaria, Pabellón 2, C1428EHA, Ciudad de Buenos Aires, Argentina.
| | - Marisa F Nicolás
- Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil.
| |
Collapse
|