1
|
Xu Y, Wang L, Guo D, Wang Y, Liu X, Sun Y, Wang R, Sun L, Jiang P, Liu Q, Wang B, Yan M, Zhao Y. Baohuoside I targets SaeR as an antivirulence strategy to disrupt MRSA biofilm formation and pathogenicity. NPJ Biofilms Microbiomes 2025; 11:45. [PMID: 40102466 PMCID: PMC11920273 DOI: 10.1038/s41522-025-00681-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 03/08/2025] [Indexed: 03/20/2025] Open
Abstract
The emergence of methicillin-resistant Staphylococcus aureus (MRSA) represents a critical global health challenge, making the SaeRS two-component system (TCS), a key regulator of S. aureus virulence, an ideal target for novel therapeutic approaches. In this study, virtual screening and thermal shift assays identified Baohuoside I (BI), a flavonol glycoside, as a potent inhibitor of the SaeR response regulator. BI significantly attenuated S. aureus pathogenicity without bactericidal effects, suppressing the expression of key virulence factors, such as hemolysin A (Hla) and Panton-Valentine leukocidin (PVL), while modulating immune evasion pathways. Additionally, BI disrupted biofilm formation, promoting the development of porous, less structured biofilms. Biochemical assays, including EMSA, CETSA, fluorescence quenching, and SPR, confirmed strong binding interactions between SaeR and BI. In vivo, BI demonstrated therapeutic efficacy in Galleria mellonella and rat MRSA models. These findings establish BI as a promising lead for nonbactericidal therapies to combat MRSA infections and mitigate resistance.
Collapse
Affiliation(s)
- Yueshan Xu
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Li Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Dongbin Guo
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yueying Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xinyao Liu
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yun Sun
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Rong Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Luanbiao Sun
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Peitong Jiang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Quan Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun, China
| | - Bingmei Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China.
| | - Ming Yan
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China.
| | - Yicheng Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun, China.
- Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China.
| |
Collapse
|
2
|
Kumar V, Chunchagatta Lakshman PK, Prasad TK, Manjunath K, Bairy S, Vasu AS, Ganavi B, Jasti S, Kamariah N. Target-based drug discovery: Applications of fluorescence techniques in high throughput and fragment-based screening. Heliyon 2024; 10:e23864. [PMID: 38226204 PMCID: PMC10788520 DOI: 10.1016/j.heliyon.2023.e23864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 01/17/2024] Open
Abstract
Target-based discovery of first-in-class therapeutics demands an in-depth understanding of the molecular mechanisms underlying human diseases. Precise measurements of cellular and biochemical activities are critical to gain mechanistic knowledge of biomolecules and their altered function in disease conditions. Such measurements enable the development of intervention strategies for preventing or treating diseases by modulation of desired molecular processes. Fluorescence-based techniques are routinely employed for accurate and robust measurements of in-vitro activity of molecular targets and for discovering novel chemical molecules that modulate the activity of molecular targets. In the current review, the authors focus on the applications of fluorescence-based high throughput screening (HTS) and fragment-based ligand discovery (FBLD) techniques such as fluorescence polarization (FP), Förster resonance energy transfer (FRET), fluorescence thermal shift assay (FTSA) and microscale thermophoresis (MST) for the discovery of chemical probe to exploring target's role in disease biology and ultimately, serve as a foundation for drug discovery. Some recent advancements in these techniques for compound library screening against important classes of drug targets, such as G-protein-coupled receptors (GPCRs) and GTPases, as well as phosphorylation- and acetylation-mediated protein-protein interactions, are discussed. Overall, this review presents a landscape of how these techniques paved the way for the discovery of small-molecule modulators and biologics against these targets for therapeutic benefits.
Collapse
Affiliation(s)
| | | | - Thazhe Kootteri Prasad
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - Kavyashree Manjunath
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - Sneha Bairy
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - Akshaya S. Vasu
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - B. Ganavi
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - Subbarao Jasti
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| | - Neelagandan Kamariah
- Centre for Chemical Biology & Therapeutics, inStem & NCBS, Bellary Road, Bangalore, 560065, India
| |
Collapse
|
3
|
Krishnan H, Ahmed S, Hubbard SR, Miller WT. Biochemical characterization of the Drosophila insulin receptor kinase and longevity-associated mutants. FASEB J 2024; 38:e23355. [PMID: 38071609 PMCID: PMC11284340 DOI: 10.1096/fj.202301948r] [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: 09/22/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023]
Abstract
Drosophila melanogaster (fruit fly) insulin receptor (D-IR) is highly homologous to the human counterpart. Like the human pathway, D-IR responds to numerous insulin-like peptides to activate cellular signals that regulate growth, development, and lipid metabolism in fruit flies. Allelic mutations in the D-IR kinase domain elevate life expectancy in fruit flies. We developed a robust heterologous expression system to express and purify wild-type and longevity-associated mutant D-IR kinase domains to investigate enzyme kinetics and substrate specificities. D-IR exhibits remarkable similarities to the human insulin receptor kinase domain but diverges in substrate preferences. We show that longevity-associated mutations reduce D-IR catalytic activity. Deletion of the unique kinase insert domain portion or mutations proximal to activating tyrosines do not influence kinase activity, suggesting their potential role in substrate recruitment and downstream signaling. Through biochemical investigations, this study enhances our comprehension of D-IR's role in Drosophila physiology, complementing genetic studies and expanding our knowledge on the catalytic functions of this conserved signaling pathway.
Collapse
Affiliation(s)
- Harini Krishnan
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Sultan Ahmed
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Stevan R. Hubbard
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Department of Veterans Affairs Medical Center, Northport, New York, USA
| |
Collapse
|
4
|
Gao X, McFadden WM, Wen X, Emanuelli A, Lorson ZC, Zheng H, Kirby KA, Sarafianos SG. Use of TSAR, Thermal Shift Analysis in R, to identify Folic Acid as a Molecule that Interacts with HIV-1 Capsid. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569293. [PMID: 38076946 PMCID: PMC10705415 DOI: 10.1101/2023.11.29.569293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Thermal shift assay (TSA) is a versatile biophysical technique for studying protein interactions. Here, we report a free, open-source software tool TSAR (Thermal Shift Analysis in R) to expedite and automate the analysis of thermal shift data derived either from individual experiments or large screens of chemical libraries. The TSAR package incorporates multiple, dynamic workflows to facilitate the analysis of TSA data and returns publication-ready graphics or processed results. Further, the package includes a graphic user interface (GUI) that enables easy use by non-programmers, aiming to simplify TSA analysis while diversifying visualization. To exemplify the utility of TSAR we screened a chemical library of vitamins to identify molecules that interact with the capsid protein (CA) of human immunodeficiency virus type 1 (HIV-1). Our data show that hexameric CA interacts with folic acid in vitro.
Collapse
Affiliation(s)
- X. Gao
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Children’s Healthcare of Atlanta, Atlanta, GA
| | - W. M. McFadden
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Children’s Healthcare of Atlanta, Atlanta, GA
| | - X. Wen
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Children’s Healthcare of Atlanta, Atlanta, GA
| | - A. Emanuelli
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Children’s Healthcare of Atlanta, Atlanta, GA
| | - Z. C. Lorson
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Children’s Healthcare of Atlanta, Atlanta, GA
| | - H. Zheng
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Children’s Healthcare of Atlanta, Atlanta, GA
| | - K. A. Kirby
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Children’s Healthcare of Atlanta, Atlanta, GA
| | - S. G. Sarafianos
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Children’s Healthcare of Atlanta, Atlanta, GA
| |
Collapse
|
5
|
Zhang C, Zhang W, Zhu S, Hu C, Che S, Wang M, Jin M, Bian N, Song W, Jiang S, Jiang Y, Hou J, Liu C, Zhou H, Wei L, Shi G, Tang Y. Bilobetin attenuates Staphylococcus aureus virulence by targeting Von Willebrand factor-binding protein and staphylocoagulase. World J Microbiol Biotechnol 2023; 39:358. [PMID: 37884743 DOI: 10.1007/s11274-023-03812-z] [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: 08/19/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
Staphylococcus aureus (S. aureus) induces a variety of infectious diseases in humans and animals and is responsible for hospital- and community-acquired infections. The aim of this study was to investigate how bilobetin, a natural compound, attenuates S. aureus virulence by inhibiting two key virulence factors, von Willebrand factor-binding protein (vWbp) and staphylocoagulase (Coa). The results showed that bilobetin inhibited Coa- or vWbp-induced coagulation without affecting S. aureus proliferation. The Western blotting and fluorescence quenching assays indicated that bilobetin did not affect the expression of vWbp and Coa but directly bound to the proteins with KA values of 1.66 × 104 L/mol and 1.04 × 104 L/mol, respectively. To gain further insight into the mechanism of interaction of bilobetin with these virulence factors, we performed molecular docking and point mutation assays, which indicated that the TYR-6 and TYR-18 residues on vWbp and the ALA-190 and ASP-189 residues on Coa were essential for the binding of bilobetin. In addition, the in vivo studies showed that bilobetin ameliorated lung tissue damage and inflammation caused by S. aureus, thereby improving the survival of mice. Furthermore, the use of bilobetin as an adjuvant in combination with vancomycin was more effective in the treatment of a mouse model of pneumonia. Taken together, bilobetin had a dual inhibitory effect on vWbp and Coa by reducing the virulence of S. aureus, suggesting that it is a viable lead compound against S. aureus infections.
Collapse
Affiliation(s)
| | - Wenyuan Zhang
- Changchun University of Chinese Medicine, Changchun, China
| | - Shuyue Zhu
- Changchun University of Chinese Medicine, Changchun, China
| | - Chunjie Hu
- Proctology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Sihua Che
- Changchun University of Chinese Medicine, Changchun, China
| | - Meiling Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - Mengli Jin
- Changchun University of Chinese Medicine, Changchun, China
| | - Nan Bian
- Changchun University of Chinese Medicine, Changchun, China
| | - Wu Song
- Changchun University of Chinese Medicine, Changchun, China
| | - Shuang Jiang
- Changchun University of Chinese Medicine, Changchun, China
| | - Yijing Jiang
- Changchun University of Chinese Medicine, Changchun, China
| | - Juan Hou
- Changchun University of Chinese Medicine, Changchun, China
| | - Chang Liu
- Changchun University of Chinese Medicine, Changchun, China
| | - Haofang Zhou
- Changchun University of Chinese Medicine, Changchun, China
| | - Lin Wei
- Changchun University of Chinese Medicine, Changchun, China.
| | - Guijun Shi
- Changchun University of Chinese Medicine, Changchun, China.
- Changchun Hospital of Traditional Chinese Medicine, Changchun, China.
| | - Yong Tang
- Changchun University of Chinese Medicine, Changchun, China.
| |
Collapse
|
6
|
Hou J, Jiang Y, Xu Y, Zhao C, Cao Y, Song W, Wang B. Inhibitory effect of bilobalide on Staphylococcus aureus von Willebrand factor-binding protein and its therapeutic effect in mice with pneumonia. J Appl Microbiol 2023; 134:lxad233. [PMID: 37833234 DOI: 10.1093/jambio/lxad233] [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: 05/18/2023] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 10/15/2023]
Abstract
AIMS Disabling bacterial virulence with small molecules has been proposed as a potential strategy to prevent bacterial pathogenicity. The von Willebrand factor-binding protein of Staphylococcus aureus was identified previously as a key virulence determinant. Our objective was to discover a von Willebrand-factor binding protein (vWbp) inhibitor distinct from the antibiotics used to prevent infections resulting from S. aureus. METHODS AND RESULTS Using coagulation assays, we found that the sesquiterpene trilactone bilobalide blocks coagulation mediated by vWbp, but has no impact on the growth of S. aureus at a concentration of 128 μg ml-1. Moreover, a mouse model of pneumonia caused by S. aureus indicated that bilobalide could attenuate S. aureus virulence in vivo. This effect is achieved not by interfering with the expression of vWbp but by binding to vWbp, as demonstrated by western blotting, thermal shift assays, and fluorescence quenching assays. Using molecular dynamic simulations and point mutagenesis analysis, we identified that the Q17A and R453A residues are key residues for the binding of bilobalide to vWbp. CONCLUSIONS Overall, we tested the ability of bilobalide to inhibit S. aureus infections by targeting vWbp and explored the potential mechanism of this activity.
Collapse
Affiliation(s)
- Juan Hou
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Yijing Jiang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Yangming Xu
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Chunhui Zhao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Yali Cao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
- Changchun University of Traditional Chinese Medicine Second Affiliated Hospital, Changchun University of Chinese Medicine, Changchun 130052, China
| | - Wu Song
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Bingmei Wang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| |
Collapse
|
7
|
Zuberek J, Warzecha M, Dobrowolski M, Modrak-Wojcik A. An intramolecular disulphide bond in human 4E-T affects its binding to eIF4E1a protein. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2023; 52:497-510. [PMID: 37798395 PMCID: PMC10618305 DOI: 10.1007/s00249-023-01684-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/14/2023] [Accepted: 09/16/2023] [Indexed: 10/07/2023]
Abstract
The cap at the 5'terminus of mRNA is a key determinant of gene expression in eukaryotic cells, which among others is required for cap dependent translation and protects mRNA from degradation. These properties of cap are mediated by several proteins. One of them is 4E-Transporter (4E-T), which plays an important role in translational repression, mRNA decay and P-bodies formation. 4E-T is also one of several proteins that interact with eukaryotic initiation factor 4E (eIF4E), a cap binding protein which is a key component of the translation initiation machinery. The molecular mechanisms underlying the interactions of these two proteins are crucial for mRNA processing. Studying the interactions between human eIF4E1a and the N-terminal fragment of 4E-T that possesses unstructured 4E-binding motifs under non-reducing conditions, we observed that 4E-T preferentially forms an intramolecular disulphide bond. This "disulphide loop" reduces affinity of 4E-T for eIF4E1a by about 300-fold. Considering that only human 4E-T possesses two cysteines located between the 4E binding motifs, we proposed that the disulphide bond may act as a switch to regulate interactions between the two proteins.
Collapse
Affiliation(s)
- Joanna Zuberek
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland.
| | - Marek Warzecha
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Mateusz Dobrowolski
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Anna Modrak-Wojcik
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| |
Collapse
|
8
|
Chen X, Hu C, Shu Z, Wang X, Zhao Y, Song W, Chen X, Jin M, Xiu Y, Guo X, Kong X, Jiang Y, Guan J, Gongga L, Wang L, Wang B. Isovanillic acid protects mice against Staphylococcus aureus by targeting vWbp and Coa. Future Microbiol 2023; 18:735-749. [PMID: 37526178 DOI: 10.2217/fmb-2022-0219] [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] [Indexed: 08/02/2023] Open
Abstract
Aim: Our primary objective was to investigate the protective effects and mechanisms of isovanillic acid in mice infected with Staphylococcus aureus Newman. Methods: In vitro coagulation assays were used to validate vWbp and Coa as inhibitory targets of isovanillic acid. The binding mechanism of isovanillic acid to vWbp and Coa was investigated using molecular docking and point mutagenesis. Importantly, a lethal pneumonia mouse model was used to assess the effect of isovanillic acid on survival and pathological injury in mice. Results & Conclusion: Isovanillic acid reduced the virulence of S. aureus by directly binding to inhibit the clotting activity of vWbp and Coa, thereby reducing lung histopathological damage and improving the survival rate in mice with pneumonia.
Collapse
Affiliation(s)
- Xiangqian Chen
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Chunjie Hu
- Changchun University of Chinese Medicine, Changchun, 130117, China
- Proctology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Zunhua Shu
- Changchun University of Chinese Medicine, Changchun, 130117, China
- The Third Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130118, China
| | - Xingye Wang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yicheng Zhao
- Changchun University of Chinese Medicine, Changchun, 130117, China
- Center for Pathogen Biology & Infectious Diseases, Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun,130021, China
| | - Wu Song
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Xiaoyu Chen
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Mengli Jin
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yang Xiu
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Xuerui Guo
- School of Pharmacy, Jilin University, Changchun, 130021, China
| | - Xiangri Kong
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yijing Jiang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jiyu Guan
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Lanzi Gongga
- Tibet University Medical College, Tibet, 850000, China
| | - Li Wang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Bingmei Wang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| |
Collapse
|
9
|
Zephyr J, Rao DN, Johnson C, Shaqra AM, Nalivaika EA, Jordan A, Kurt Yilmaz N, Ali A, Schiffer CA. Allosteric quinoxaline-based inhibitors of the flavivirus NS2B/NS3 protease. Bioorg Chem 2023; 131:106269. [PMID: 36446201 PMCID: PMC10155214 DOI: 10.1016/j.bioorg.2022.106269] [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: 09/16/2022] [Revised: 10/27/2022] [Accepted: 11/06/2022] [Indexed: 11/20/2022]
Abstract
Viruses from the Flavivirus genus infect millions of people worldwide and cause severe diseases, including recent epidemics of dengue virus (DENV), and Zika virus (ZIKV). There is currently no antiviral treatment against flavivirus infections, despite considerable efforts to develop inhibitors against essential viral enzymes including NS2B/NS3 protease. Targeting the flavivirus NS2B/NS3 protease proved to be challenging because of the conformational dynamics, topology, and electrostatic properties of the active site. Here, we report the identification of quinoxaline-based allosteric inhibitors by fragment-based drug discovery approach as a promising new drug-like scaffold to target the NS2B/NS3 protease. Enzymatic assays and mutational analysis of the allosteric site in ZIKV NS2B/NS3 protease support noncompetitive inhibition mechanism as well as engineered DENV protease construct indicating the compounds likely compete with the NS2B cofactor for binding to the protease domain. Furthermore, antiviral activity confirmed the therapeutic potential of this new inhibitor scaffold.
Collapse
Affiliation(s)
- Jacqueto Zephyr
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Desaboini Nageswara Rao
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Colby Johnson
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Ala M Shaqra
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Ellen A Nalivaika
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Aria Jordan
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States.
| | - Akbar Ali
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States.
| | - Celia A Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States.
| |
Collapse
|
10
|
Hinokiflavone Attenuates the Virulence of Methicillin-Resistant Staphylococcus aureus by Targeting Caseinolytic Protease P. Antimicrob Agents Chemother 2022; 66:e0024022. [PMID: 35862746 PMCID: PMC9380526 DOI: 10.1128/aac.00240-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Drug-resistant bacteria was the third leading cause of death worldwide in 2019, which sounds like a cautionary note for global public health. Therefore, developing novel strategies to combat Methicillin-resistant Staphylococcus aureus (MRSA) infections is the need of the hour. Caseinolytic protease P (ClpP) represents pivotal microbial degradation machinery in MRSA involved in bacterial homeostasis and pathogenicity, considered an ideal target for combating S. aureus infections. Herein, we identified a natural compound, hinokiflavone, that inhibited the activity of ClpP of MRSA strain USA300 with an IC50 of 34.36 μg/mL. Further assays showed that hinokiflavone reduced the virulence of S. aureus by inhibiting multiple virulence factors expression. Results obtained from cellular thermal transfer assay (CETSA), thermal shift assay (TSA), local surface plasmon resonance (LSPR) and molecular docking (MD) assay enunciated that hinokiflavone directly bonded to ClpP with confirmed docking sites, including SER-22, LYS-26 and ARG-28. In vivo, the evaluation of anti-infective activity showed that hinokiflavone in combination with vancomycin effectively protected mice from MRSA-induced fatal pneumonia, which was more potent than vancomycin alone. As mentioned above, hinokiflavone, as an inhibitor of ClpP, could be further developed into a promising adjuvant against S. aureus infections.
Collapse
|
11
|
Tan F, Xu J. Validation of the solution structure of dimerization domain of PRC1. PLoS One 2022; 17:e0270572. [PMID: 35930764 PMCID: PMC9355583 DOI: 10.1371/journal.pone.0270572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/13/2022] [Indexed: 11/18/2022] Open
Abstract
Cell-cycle dependent proteins are indispensible for the accurate division of cells, a group of proteins called Microtubule-associated proteins (MAPs) are important to cell division as it bind microtubules and participate with other co-factors to form the spindle midbody, which works as the workhorse of cell-division. PRC1 is a distinguishing member of MAPs, as it is a human MAP and works as the key in mediating daughter cell segregation in ana-phase and telo-phase. The physiological significance of PRC1 calls for a high resolution three-dimensional structure. The crystal structure of PRC1 was published but has low resolution (>3 Å) and incomplete sidechains, placing hurdles to understanding the structure-function relationships of PRC1, therefore, we determined the high-resolution solution structure of PRC1’s dimerization domain using NMR spectroscopy. Significant differences between the crystal structure and the solution structure can be observed, the main differences center around the N terminus and the end of the alpha-Helix H2. Furthermore, detailed structure analyses revealed that the hydrophobic core packing of the solution and crystal structures are also different. To validate the solution structure, we used Hydrogen-deuterium exchange experiments that address the structural discrepancies between the crystal and solution structure; we also generated mutants that are key to the differences in the crystal and solution structures, measuring its structural or thermal stability by NMR spectroscopy and Fluorescence Thermal Shift Assays. These results suggest that N terminal residues are key to the integrity of the whole protein, and the solution structure of the dimerization domain better reflects the conformation PRC1 adopted in solution conditions.
Collapse
Affiliation(s)
- Fei Tan
- Peking University, Beijing, China
- * E-mail:
| | - Jin Xu
- Peking University, Beijing, China
| |
Collapse
|
12
|
Cheng C, Liu M, Gao X, Wu D, Pu M, Ma J, Quinn RJ, Xiao Z, Liu Z. Identifying New Ligands for JNK3 by Fluorescence Thermal Shift Assays and Native Mass Spectrometry. ACS OMEGA 2022; 7:13925-13931. [PMID: 35559183 PMCID: PMC9088906 DOI: 10.1021/acsomega.2c00340] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/05/2022] [Indexed: 06/15/2023]
Abstract
The c-Jun N-terminal kinases (JNKs) are evolutionary highly conserved serine/threonine kinases. Numerous findings suggest that JNK3 is involved in the pathogenesis of neurodegenerative diseases, so the inhibition of JNK3 may be a potential therapeutic intervention. The identification of novel compounds with promising pharmacological properties still represents a challenge. Fluorescence thermal shift screening of a chemically diversified lead-like scaffold library of 2024 pure compounds led to the initial identification of seven JNK3 binding hits, which were classified into four scaffold groups according to their chemical structures. Native mass spectrometry validated the interaction of 4 out of the 7 hits with JNK3. Binding geometries and interactions of the top 2 hits were evaluated by docking into a JNK3 crystal structure. Hit 5 had a K d of 21 μM with JNK3 suggested scaffold 5-(phenylamino)-1H-1,2,3-triazole-4-carboxamide as a novel and selective JNK3 binder.
Collapse
Affiliation(s)
- Chongyun Cheng
- National
Laboratory of Biomacromolecules, Institute
of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
- Monash
Biomedicine Discovery Institute, Monash
University, Melbourne, Victoria 3800, Australia
| | - Miaomiao Liu
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
| | - Xiaoqin Gao
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
| | - Dong Wu
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Mengchen Pu
- National
Laboratory of Biomacromolecules, Institute
of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Ma
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
| | - Ronald J. Quinn
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
| | - Zhicheng Xiao
- Monash
Biomedicine Discovery Institute, Monash
University, Melbourne, Victoria 3800, Australia
- Kunming
Medical College, Kunming, Yunnan 650031, China
| | - Zhijie Liu
- National
Laboratory of Biomacromolecules, Institute
of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
- Kunming
Medical College, Kunming, Yunnan 650031, China
| |
Collapse
|
13
|
Monteagudo-Cascales E, Santero E, Canosa I. The Regulatory Hierarchy Following Signal Integration by the CbrAB Two-Component System: Diversity of Responses and Functions. Genes (Basel) 2022; 13:genes13020375. [PMID: 35205417 PMCID: PMC8871633 DOI: 10.3390/genes13020375] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
CbrAB is a two-component system, unique to bacteria of the family Pseudomonaceae, capable of integrating signals and involved in a multitude of physiological processes that allow bacterial adaptation to a wide variety of varying environmental conditions. This regulatory system provides a great metabolic versatility that results in excellent adaptability and metabolic optimization. The two-component system (TCS) CbrA-CbrB is on top of a hierarchical regulatory cascade and interacts with other regulatory systems at different levels, resulting in a robust output. Among the regulatory systems found at the same or lower levels of CbrAB are the NtrBC nitrogen availability adaptation system, the Crc/Hfq carbon catabolite repression cascade in Pseudomonas, or interactions with the GacSA TCS or alternative sigma ECF factor, such as SigX. The interplay between regulatory mechanisms controls a number of physiological processes that intervene in important aspects of bacterial adaptation and survival. These include the hierarchy in the use of carbon sources, virulence or resistance to antibiotics, stress response or definition of the bacterial lifestyle. The multiple actions of the CbrAB TCS result in an important competitive advantage.
Collapse
Affiliation(s)
| | - Eduardo Santero
- Departamento de Biología Molecular e Ingeniería Bioquímica, Universidad Pablo de Olavide, Centro Andaluz de Biología del Desarrollo, CSIC, Junta de Andalucía, 41013 Seville, Spain;
| | - Inés Canosa
- Departamento de Biología Molecular e Ingeniería Bioquímica, Universidad Pablo de Olavide, Centro Andaluz de Biología del Desarrollo, CSIC, Junta de Andalucía, 41013 Seville, Spain;
- Correspondence: ; Tel.: +34-954349052
| |
Collapse
|
14
|
Xiang H, Yang P, Wang L, Li J, Wang T, Xue J, Wang D, Ma H. Isovitexin Is a Direct Inhibitor of Staphylococcus aureus Coagulase. J Microbiol Biotechnol 2021; 31:1350-1357. [PMID: 34409949 PMCID: PMC9706020 DOI: 10.4014/jmb.2105.05013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/28/2021] [Accepted: 08/17/2021] [Indexed: 12/15/2022]
Abstract
Staphylococcus aureus (S. aureus) is a major pathogen that causes human pneumonia, leading to significant morbidity and mortality. S. aureus coagulase (Coa) triggers the polymerization of fibrin by activating host prothrombin, which then converts fibrinogen to fibrin and contributes to S. aureus pathogenesis and persistent infection. In our research, we demonstrate that isovitexin, an active traditional Chinese medicine component, can inhibit the coagulase activity of Coa but does not interfere with the growth of S. aureus. Furthermore, we show through thermal shift and fluorescence quenching assays that isovitexin directly binds to Coa. Dynamic simulation and structure-activity relationship analyses suggest that V191 and P268 are key amino acid residues responsible for the binding of isovitexin to Coa. Taken together, these data indicate that isovitexin is a direct Coa inhibitor and a promising candidate for drug development against S. aureus infection.
Collapse
Affiliation(s)
- Hua Xiang
- College of Animal Medicine, Jilin Agricultural University, Changchun 130118, P.R. China,College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, P.R. China,The Key Laboratory of New Veterinary Drug Research and Development of Jilin Province, Jilin Agricultural University, Changchun 130118, P.R. China
| | - Panpan Yang
- College of Basic Medical Science, Jilin University, Changchun 130012, P.R. China
| | - Li Wang
- College of Animal Science, Jilin University, Changchun 130062, P.R. China
| | - Jiaxin Li
- College of Animal Science, Jilin University, Changchun 130062, P.R. China
| | - Tiedong Wang
- College of Animal Science, Jilin University, Changchun 130062, P.R. China
| | - Junze Xue
- College of Animal Medicine, Jilin Agricultural University, Changchun 130118, P.R. China,College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, P.R. China,The Key Laboratory of New Veterinary Drug Research and Development of Jilin Province, Jilin Agricultural University, Changchun 130118, P.R. China
| | - Dacheng Wang
- College of Animal Science, Jilin University, Changchun 130062, P.R. China,Corresponding author Phone: +86-431-84532812 E-mail:
| | - Hongxia Ma
- College of Animal Medicine, Jilin Agricultural University, Changchun 130118, P.R. China,College of Life Science, Jilin Agricultural University, Changchun 130118, P.R. China,The Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, Jilin Agricultural University, Changchun 130118, P.R. China,Corresponding author Phone: +86-431-84532812 E-mail:
| |
Collapse
|
15
|
Toward Chemical Validation of Leishmania infantum Ribose 5-Phosphate Isomerase as a Drug Target. Antimicrob Agents Chemother 2021; 65:e0189220. [PMID: 33875438 DOI: 10.1128/aac.01892-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neglected tropical diseases caused by kinetoplastid parasites (Trypanosoma brucei, Trypanosoma cruzi, and Leishmania spp.) place a significant health and economic burden on developing nations worldwide. Current therapies are largely outdated, inadequate, and face mounting drug resistance from the causative parasites. Thus, there is an urgent need for drug discovery and development. Target-led drug discovery approaches have focused on the identification of parasite enzymes catalyzing essential biochemical processes, which significantly differ from equivalent proteins found in humans, thereby providing potentially exploitable therapeutic windows. One such target is ribose 5-phosphate isomerase B (RpiB), an enzyme involved in the nonoxidative branch of the pentose phosphate pathway, which catalyzes the interconversion of d-ribose 5-phosphate and d-ribulose 5-phosphate. Although protozoan RpiB has been the focus of numerous targeted studies, compounds capable of selectively inhibiting this parasite enzyme have not been identified. Here, we present the results of a fragment library screening against Leishmania infantum RpiB (LiRpiB), performed using thermal shift analysis. Hit fragments were shown to be effective inhibitors of LiRpiB in activity assays, and several fragments were capable of selectively inhibiting parasite growth in vitro. These results support the identification of LiRpiB as a validated therapeutic target. The X-ray crystal structure of apo LiRpiB was also solved, permitting docking studies to assess how hit fragments might interact with LiRpiB to inhibit its activity. Overall, this work will guide structure-based development of LiRpiB inhibitors as antileishmanial agents.
Collapse
|
16
|
Torres‐Ocampo AP, Özden C, Hommer A, Gardella A, Lapinskas E, Samkutty A, Esposito E, Garman SC, Stratton MM. Characterization of CaMKIIα holoenzyme stability. Protein Sci 2020; 29:1524-1534. [PMID: 32282091 PMCID: PMC7255518 DOI: 10.1002/pro.3869] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/29/2022]
Abstract
Ca2+ /calmodulin-dependent protein kinase II (CaMKII) is a Ser/Thr kinase necessary for long-term memory formation and other Ca2+ -dependent signaling cascades such as fertilization. Here, we investigated the stability of CaMKIIα using a combination of differential scanning calorimetry (DSC), X-ray crystallography, and mass photometry (MP). The kinase domain has a low thermal stability (apparent Tm = 36°C), which is slightly stabilized by ATP/MgCl2 binding (apparent Tm = 40°C) and significantly stabilized by regulatory segment binding (apparent Tm = 60°C). We crystallized the kinase domain of CaMKII bound to p-coumaric acid in the active site. This structure reveals solvent-exposed hydrophobic residues in the substrate-binding pocket, which are normally buried in the autoinhibited structure when the regulatory segment is present. This likely accounts for the large stabilization that we observe in DSC measurements comparing the kinase alone with the kinase plus regulatory segment. The hub domain alone is extremely stable (apparent Tm ~ 90°C), and the holoenzyme structure has multiple unfolding transitions ranging from ~60°C to 100°C. Using MP, we compared a CaMKIIα holoenzyme with different variable linker regions and determined that the dissociation of both these holoenzymes occurs at a higher concentration (is less stable) compared with the hub domain alone. We conclude that within the context of the holoenzyme structure, the kinase domain is stabilized, whereas the hub domain is destabilized. These data support a model where domains within the holoenzyme interact.
Collapse
Affiliation(s)
- Ana P. Torres‐Ocampo
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
- Molecular and Cellular Biology Graduate ProgramUniversity of MassachusettsAmherstMassachusettsUSA
| | - Can Özden
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
- Molecular and Cellular Biology Graduate ProgramUniversity of MassachusettsAmherstMassachusettsUSA
| | - Alexandra Hommer
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Anne Gardella
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Emily Lapinskas
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Alfred Samkutty
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | | | - Scott C Garman
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Margaret M Stratton
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
- Molecular and Cellular Biology Graduate ProgramUniversity of MassachusettsAmherstMassachusettsUSA
| |
Collapse
|
17
|
Targeting staphylocoagulase with isoquercitrin protects mice from Staphylococcus aureus-induced pneumonia. Appl Microbiol Biotechnol 2020; 104:3909-3919. [PMID: 32130467 DOI: 10.1007/s00253-020-10486-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 10/24/2022]
Abstract
Staphylocoagulase (Coa) is a virulence factor of Staphylococcus aureus (S. aureus) that promotes blood coagulation by activating prothrombin to convert fibrinogen to fibrin. Coa plays a crucial role in disease pathogenesis and is a promising target for the treatment of S. aureus infections. Here, we identified that isoquercitrin, a natural flavonol compound, can markedly reduce the activity of Coa at concentrations that have no effect on bacterial growth. Mechanistic studies employing molecular dynamics simulation revealed that isoquercitrin binds to Coa by interacting with Asp-181 and Tyr-188, thereby affecting the binding of Coa to prothrombin. Importantly, in vivo studies showed that isoquercitrin treatment significantly reduced the bacterial burden, pathological damage, and inflammation of lung tissue and improved the percentage of survival of mice infected with S. aureus Newman strain. These data suggest that isoquercitrin is a promising inhibitor of Coa that can be used for the development of therapeutic drugs to combat S. aureus infections.Key Points• Staphylocoagulase plays a key role in the pathogenesis of S. aureus infection.• We identified that isoquercitrin is a direct inhibitor of staphylocoagulase.• Isoquercitrin treatment can significantly attenuate S. aureus virulence in vivo.
Collapse
|
18
|
O’Byrne SN, Scott JW, Pilotte JR, Santiago ADS, Langendorf CG, Oakhill JS, Eduful BJ, Couñago RM, Wells CI, Zuercher WJ, Willson TM, Drewry DH. In Depth Analysis of Kinase Cross Screening Data to Identify CAMKK2 Inhibitory Scaffolds. Molecules 2020; 25:E325. [PMID: 31941153 PMCID: PMC7024175 DOI: 10.3390/molecules25020325] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/25/2022] Open
Abstract
The calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) activates CAMK1, CAMK4, AMPK, and AKT, leading to numerous physiological responses. The deregulation of CAMKK2 is linked to several diseases, suggesting the utility of CAMKK2 inhibitors for oncological, metabolic and inflammatory indications. In this work, we demonstrate that STO-609, frequently described as a selective inhibitor for CAMKK2, potently inhibits a significant number of other kinases. Through an analysis of literature and public databases, we have identified other potent CAMKK2 inhibitors and verified their activities in differential scanning fluorimetry and enzyme inhibition assays. These inhibitors are potential starting points for the development of selective CAMKK2 inhibitors and will lead to tools that delineate the roles of this kinase in disease biology.
Collapse
Affiliation(s)
- Sean N. O’Byrne
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - John W. Scott
- St Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia; (J.W.S.); (C.G.L.); (J.S.O.)
- Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring Street, Melbourne 3000, Australia
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville 3052, Australia
| | - Joseph R. Pilotte
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - André da S. Santiago
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas SP 13083-875, Brazil; (A.d.S.S.); (R.M.C.)
- Structural Genomics Consortium, Departamento de Genética e Evolução, Instituto de Biologia, UNICAMP, Campinas SP 13083-886, Brazil
| | - Christopher G. Langendorf
- St Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia; (J.W.S.); (C.G.L.); (J.S.O.)
| | - Jonathan S. Oakhill
- St Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia; (J.W.S.); (C.G.L.); (J.S.O.)
- Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring Street, Melbourne 3000, Australia
| | - Benjamin J. Eduful
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - Rafael M. Couñago
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas SP 13083-875, Brazil; (A.d.S.S.); (R.M.C.)
- Structural Genomics Consortium, Departamento de Genética e Evolução, Instituto de Biologia, UNICAMP, Campinas SP 13083-886, Brazil
| | - Carrow I. Wells
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - William J. Zuercher
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - Timothy M. Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - David H. Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| |
Collapse
|
19
|
Wilson CG, Arkin MR. Screening and biophysics in small molecule discovery. SMALL MOLECULE DRUG DISCOVERY 2020:127-161. [DOI: 10.1016/b978-0-12-818349-6.00005-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
20
|
Mishra RK, Clutter MR, Blyth GT, Kosciuczuk EM, Blackburn AZ, Beauchamp EM, Schiltz GE, Platanias LC. Discovery of novel Mnk inhibitors using mutation-based induced-fit virtual high-throughput screening. Chem Biol Drug Des 2019; 94:1813-1823. [PMID: 31260185 DOI: 10.1111/cbdd.13585] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022]
Abstract
Mnk kinases (Mnk1 and 2) are downstream effectors of Map kinase pathways and regulate phosphorylation of eukaryotic initiation factor 4E. Engagement of the Mnk pathway is critical in acute myeloid leukemia (AML) leukemogenesis and Mnk inhibitors have potent antileukemic properties in vitro and in vivo, suggesting that targeting Mnk kinases may provide a novel approach for treating AML. Here, we report the development and application of a mutation-based induced-fit in silico screen to identify novel Mnk inhibitors. The Mnk1 structure was modeled by temporarily mutating an amino acid that obstructs the ATP-binding site in the Mnk1 crystal structure while carrying out docking simulations of known inhibitors. The hit compounds display activity in Mnk biochemical and cellular assays, including acute myeloid leukemia progenitors. This approach will enable further rational structure-based drug design of new Mnk inhibitors and potentially novel ways of therapeutically targeting this kinase.
Collapse
Affiliation(s)
- Rama K Mishra
- The Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, USA.,Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Matthew R Clutter
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Gavin T Blyth
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ewa M Kosciuczuk
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Amy Z Blackburn
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elspeth M Beauchamp
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Gary E Schiltz
- The Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, USA.,Department of Pharmacology, Northwestern University, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
21
|
Wang L, Li B, Si X, Liu X, Deng X, Niu X, Jin Y, Wang D, Wang J. Quercetin protects rats from catheter-related Staphylococcus aureus infections by inhibiting coagulase activity. J Cell Mol Med 2019; 23:4808-4818. [PMID: 31094081 PMCID: PMC6584481 DOI: 10.1111/jcmm.14371] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 03/18/2019] [Accepted: 04/22/2019] [Indexed: 11/30/2022] Open
Abstract
Coagulase (Coa) activity is essential for the virulence of Staphylococcus aureus (S aureus), one of the most important pathogenic bacteria leading to catheter‐related bloodstream infections (CRBSI). We have demonstrated that the mutation of coagulase improved outcomes in disease models of S aureus CRBSI, suggesting that targeting Coa may represent a novel antiinfective strategy for CRBSI. Here, we found that quercetin, a natural compound that does not affect S aureus viability, could inhibit Coa activity. Chemical biological analysis revealed that the direct engagement of quercetin with the active site (residues Tyr187, Leu221 and His228) of Coa inhibited its activity. Furthermore, treatment with quercetin reduced the retention of bacteria on catheter surfaces, decreased the bacterial load in the kidneys and alleviated kidney abscesses in vivo. These data suggest that antiinfective therapy targeting Coa with quercetin may represent a novel strategy and provide a new leading compound with which to combat bacterial infections.
Collapse
Affiliation(s)
- Lin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - BangBang Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China.,College of Animal Science, Jilin University, Changchun, Jilin, China.,The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaosa Si
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xuming Deng
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xiaodi Niu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yingli Jin
- Department of Pharmacology, College of Basic Medical Science, Jilin University, Changchun, Jilin, China
| | - Dacheng Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China.,College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Jianfeng Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| |
Collapse
|
22
|
Deibler KK, Schiltz GE, Clutter MR, Mishra RK, Vagadia PP, O’Connor M, George MD, Gordon R, Fowler G, Bergan R, Scheidt KA. Synthesis and Biological Evaluation of 3-Arylindazoles as Selective MEK4 Inhibitors. ChemMedChem 2019; 14:615-620. [PMID: 30707493 PMCID: PMC6476181 DOI: 10.1002/cmdc.201900019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 01/30/2019] [Indexed: 01/19/2023]
Abstract
Herein we report the discovery of a novel series of highly potent and selective mitogen-activated protein kinase kinase 4 (MEK4) inhibitors. MEK4 is an upstream kinase in MAPK signaling pathways that phosphorylates p38 MAPK and JNK in response to mitogenic and cellular stress queues. MEK4 is overexpressed and induces metastasis in advanced prostate cancer lesions. However, the value of MEK4 as an oncology target has not been pharmacologically validated because selective chemical probes targeting MEK4 have not been developed. Optimization of this series via structure-activity relationships and molecular modeling led to the identification of compound 6 ff (4-(6-fluoro-2H-indazol-3-yl)benzoic acid), a highly potent and selective MEK4 inhibitor. This series of inhibitors is the first of its kind in both activity and selectivity and will be useful in further defining the role of MEK4 in prostate and other cancers.
Collapse
Affiliation(s)
- Kristine K. Deibler
- Department of Chemistry, Northwestern University, Evanston, 60208, Illinois, USA,
| | - Gary E. Schiltz
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois, 60208, USA
- Department of Pharmacology, Northwestern University, Chicago, 60611, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| | - Matthew R. Clutter
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
- Chemistry of Life Process Institute, Northwestern University, Evanston, 60208, Illinois, USA
- Department of Molecular Biosciences, Northwestern University, Evanston, 60208, Illinois, USA
| | - Rama K. Mishra
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois, 60208, USA
- Department of Pharmacology, Northwestern University, Chicago, 60611, Illinois, USA
| | - Purav P. Vagadia
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois, 60208, USA
| | - Matthew O’Connor
- Department of Chemistry, Northwestern University, Evanston, 60208, Illinois, USA,
| | - Mariam Donny George
- Chemistry of Life Process Institute, Northwestern University, Evanston, 60208, Illinois, USA
| | - Ryan Gordon
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Graham Fowler
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Raymond Bergan
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Karl A. Scheidt
- Department of Chemistry, Northwestern University, Evanston, 60208, Illinois, USA,
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois, 60208, USA
- Department of Pharmacology, Northwestern University, Chicago, 60611, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
- Chemistry of Life Process Institute, Northwestern University, Evanston, 60208, Illinois, USA
| |
Collapse
|
23
|
Li B, Jin Y, Xiang H, Mu D, Yang P, Li X, Zhong L, Cao J, Xu D, Gong Q, Wang T, Wang L, Wang D. An Inhibitory Effect of Dryocrassin ABBA on Staphylococcus aureus vWbp That Protects Mice From Pneumonia. Front Microbiol 2019; 10:7. [PMID: 30728809 PMCID: PMC6351477 DOI: 10.3389/fmicb.2019.00007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/07/2019] [Indexed: 01/18/2023] Open
Abstract
Von Willebrand factor-binding protein (vWbp), secreted by Staphylococcus aureus (S. aureus), can activate host prothrombin, convert fibrinogen to fibrin clots, induce blood clotting, and contribute to pathophysiology of S. aureus-related diseases, including infective endocarditis, staphylococcal sepsis and pneumonia. Therefore, vWbp is an promising drug target in the treatment of S. aureus-related infections. Here, we report that dryocrassin ABBA (ABBA), a natural compound derived from Dryopteris crassirhizoma, can significantly inhibit the coagulase activity of vWbp in vitro by directly interacting with vWbp without killing the bacteria or inhibiting the expression of the vWbp. Using molecular dynamics simulations, we demonstrate that ABBA binds to the "central cavity" in the elbow of vWbp by interacting with Arg-70, His-71, Ala-72, Gly-73, Tyr-74, Glu-75, Tyr-83, and Gln-87 in vWbp, thus interfering with the binding of vWbp to prothrombin. Furthermore, in vivo studies demonstrated that ABBA can attenuate injury and inflammation of mouse lung tissues caused by S. aureus and increase survival of mice. Together these findings indicate that ABBA is a promising lead drug for the treatment of S. aureus-related infections. This is the first report of potential inhibitor which inhibit the coagulase activity of vWbp by directly interacting with vWbp.
Collapse
Affiliation(s)
- Bangbang Li
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yingli Jin
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Hua Xiang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Dan Mu
- College of Animal Sciences, Jilin University, Changchun, China
| | - Panpan Yang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xianmei Li
- College of Animal Sciences, Jilin University, Changchun, China
| | - Ling Zhong
- College of Animal Sciences, Jilin University, Changchun, China
| | - Junjie Cao
- College of Animal Sciences, Jilin University, Changchun, China
| | - Dan Xu
- Shen Yang Weijia Animal Husbandry Company Limited, Shenyang, China
| | - Qian Gong
- College of Humanities & Sciences of Northeast Normal University, Changchun, China
| | - Tiedong Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Lin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Dacheng Wang
- College of Animal Sciences, Jilin University, Changchun, China
| |
Collapse
|
24
|
Wong EL, Nawrotzky E, Arkona C, Kim BG, Beligny S, Wang X, Wagner S, Lisurek M, Carstanjen D, Rademann J. The transcription factor STAT5 catalyzes Mannich ligation reactions yielding inhibitors of leukemic cell proliferation. Nat Commun 2019; 10:66. [PMID: 30622248 PMCID: PMC6325109 DOI: 10.1038/s41467-018-07923-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022] Open
Abstract
Protein-templated fragment ligations have been established as a powerful method for the assembly and detection of optimized protein ligands. Initially developed for reversible ligations, the method has been expanded to irreversible reactions enabling the formation of super-additive fragment combinations. Here, protein-induced Mannich ligations are discovered as a biocatalytic reaction furnishing inhibitors of the transcription factor STAT5. STAT5 protein catalyzes multicomponent reactions of a phosphate mimetic, formaldehyde, and 1H-tetrazoles yielding protein ligands with greatly increased binding affinity and ligand efficiency. Reactions are induced under physiological conditions selectively by native STAT5 but not by other proteins. Formation of ligation products and (auto-)inhibition of the reaction are quantified and the mechanism is investigated. Inhibitors assembled by STAT5 block specifically the phosphorylation of this protein in a cellular model of acute myeloid leukemia (AML), DNA-binding of STAT5 dimers, expression of downstream targets of the transcription factor, and the proliferation of cancer cells in mice. The oncogene STAT5 is involved in cancer cell proliferation. Here, the authors use STAT5 protein to assemble its own small molecule inhibitors via Mannich ligation (three-component-reactions) and show that the resultant ligands can inhibit the proliferation of cancer cells in a mouse model.
Collapse
Affiliation(s)
- Ee Lin Wong
- Department of Biology, Chemistry and Pharmacy, Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195, Berlin, Germany
| | - Eric Nawrotzky
- Department of Biology, Chemistry and Pharmacy, Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195, Berlin, Germany
| | - Christoph Arkona
- Department of Biology, Chemistry and Pharmacy, Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195, Berlin, Germany
| | - Boo Geun Kim
- Department of Medicinal Chemistry, Leibniz Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Samuel Beligny
- Department of Medicinal Chemistry, Leibniz Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Xinning Wang
- Department of Biology, Chemistry and Pharmacy, Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195, Berlin, Germany
| | - Stefan Wagner
- Department of Biology, Chemistry and Pharmacy, Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195, Berlin, Germany
| | - Michael Lisurek
- Department of Medicinal Chemistry, Leibniz Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Dirk Carstanjen
- Department of Medicinal Chemistry, Leibniz Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Jörg Rademann
- Department of Biology, Chemistry and Pharmacy, Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195, Berlin, Germany. .,Department of Medicinal Chemistry, Leibniz Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125, Berlin, Germany.
| |
Collapse
|
25
|
Entropic contribution to enhanced thermal stability in the thermostable P450 CYP119. Proc Natl Acad Sci U S A 2018; 115:E10049-E10058. [PMID: 30297413 PMCID: PMC6205451 DOI: 10.1073/pnas.1807473115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The enhanced thermostability of thermophilic proteins with respect to their mesophilic counterparts is often attributed to the enthalpy effect, arising from strong interactions between protein residues. Intuitively, these strong interresidue interactions will rigidify the biomolecules. However, the present work utilizing neutron scattering and solution NMR spectroscopy measurements demonstrates a contrary example that the thermophilic cytochrome P450, CYP119, is much more flexible than its mesophilic counterpart, CYP101A1, something which is not apparent just from structural comparison of the two proteins. A mechanism to explain this apparent contradiction is that higher flexibility in the folded state of CYP119 increases its conformational entropy and thereby reduces the entropy gain during denaturation, which will increase the free energy needed for unfolding and thus stabilize the protein. This scenario is supported by thermodynamic data on the temperature dependence of unfolding free energy, which shows a significant entropic contribution to the thermostability of CYP119 and lends an added dimension to enhanced stability, previously attributed only to presence of aromatic stacking interactions and salt bridge networks. Our experimental data also support the notion that highly thermophilic P450s such as CYP119 may use a mechanism that partitions flexibility differently from mesophilic P450s between ligand binding and thermal stability.
Collapse
|
26
|
Xu L, Gordon R, Farmer R, Pattanayak A, Binkowski A, Huang X, Avram M, Krishna S, Voll E, Pavese J, Chavez J, Bruce J, Mazar A, Nibbs A, Anderson W, Li L, Jovanovic B, Pruell S, Valsecchi M, Francia G, Betori R, Scheidt K, Bergan R. Precision therapeutic targeting of human cancer cell motility. Nat Commun 2018; 9:2454. [PMID: 29934502 PMCID: PMC6014988 DOI: 10.1038/s41467-018-04465-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 05/02/2018] [Indexed: 12/12/2022] Open
Abstract
Increased cancer cell motility constitutes a root cause of end organ destruction and mortality, but its complex regulation represents a barrier to precision targeting. We use the unique characteristics of small molecules to probe and selectively modulate cell motility. By coupling efficient chemical synthesis routes to multiple upfront in parallel phenotypic screens, we identify that KBU2046 inhibits cell motility and cell invasion in vitro. Across three different murine models of human prostate and breast cancer, KBU2046 inhibits metastasis, decreases bone destruction, and prolongs survival at nanomolar blood concentrations after oral administration. Comprehensive molecular, cellular and systemic-level assays all support a high level of selectivity. KBU2046 binds chaperone heterocomplexes, selectively alters binding of client proteins that regulate motility, and lacks all the hallmarks of classical chaperone inhibitors, including toxicity. We identify a unique cell motility regulatory mechanism and synthesize a targeted therapeutic, providing a platform to pursue studies in humans. In this study, the authors identify and validate a halogen-substituted isoflavanone able to inhibit prostate cancer cell motility, invasion and metastasis in vitro and in vivo. They demonstrate its ability to selectively inhibit activation of client proteins that stimulate cell motility.
Collapse
Affiliation(s)
- Li Xu
- Department of Medicine, Northwestern University, Chicago, IL, 60611, USA.,Department of Gastroenterology, Xiang'an Hospital of Xiamen University, Fujian, 361101, Xiamen, China
| | - Ryan Gordon
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Rebecca Farmer
- Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Abhinandan Pattanayak
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Andrew Binkowski
- Department of Computer Science, University of Chicago, Chicago, IL, 60637, USA
| | - Xiaoke Huang
- Department of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Michael Avram
- Department of Anesthesiology, Northwestern University, Chicago, IL, 60611, USA
| | - Sankar Krishna
- Department of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Eric Voll
- Department of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Janet Pavese
- Department of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Juan Chavez
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - James Bruce
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Andrew Mazar
- Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Antoinette Nibbs
- Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Wayne Anderson
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, Chicago, IL, 60611, USA
| | - Lin Li
- Department of Pathology, Northwestern University, Chicago, IL, 60611, USA
| | - Borko Jovanovic
- Department of Preventive Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sean Pruell
- Department of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Matias Valsecchi
- Department of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Rick Betori
- Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Karl Scheidt
- Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Raymond Bergan
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
27
|
Singh J, Khan MI, Singh Yadav SP, Srivastava A, Sinha KK, Ashish, Das P, Kundu B. L-Asparaginase of Leishmania donovani: Metabolic target and its role in Amphotericin B resistance. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2017; 7:337-349. [PMID: 28988014 PMCID: PMC5633258 DOI: 10.1016/j.ijpddr.2017.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/22/2017] [Accepted: 09/21/2017] [Indexed: 01/12/2023]
Abstract
Emergence of Amphotericin B (AmB) resistant Leishmania donovani has posed major therapeutic challenge against the parasite. Consequently, combination therapy aimed at multiple molecular targets, based on proteome wise network analysis has been recommended. In this regard we had earlier identified and proposed L-asparaginase of Leishmania donovani (LdAI) as a crucial metabolic target. Here we report that both LdAI overexpressing axenic amastigote and promastigote forms of L. donovani survives better when challenged with AmB as compared to wild type strain. Conversely, qRT-PCR analysis showed an upregulation of LdAI in both forms upon AmB treatment. Our data demonstrates the importance of LdAI in imparting immediate protective response to the parasite upon AmB treatment. In the absence of structural and functional information, we modeled LdAI and validated its solution structure through small angle X-ray scattering (SAXS) analysis. We identified its specific inhibitors through ligand and structure-based approach and characterized their effects on enzymatic properties (Km, Vmax, Kcat) of LdAI. We show that in presence of two of the inhibitors L1 and L2, the survival of L. donovani is compromised whereas overexpression of LdAI in these cells restores viability. Taken together, our results conclusively prove that LdAI is a crucial metabolic enzyme conferring early counter measure against AmB treatment by Leishmania.
Collapse
Affiliation(s)
- Jasdeep Singh
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Mohd Imran Khan
- National Institute of Pharmaceutical Education & Research, EPIP Complex, Hajipur, Vaishali 844102, India
| | - Shiv Pratap Singh Yadav
- The Council of Scientific and Industrial Research Institute of Microbial Technology, Chandigarh, India
| | - Ankit Srivastava
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Kislay K Sinha
- National Institute of Pharmaceutical Education & Research, EPIP Complex, Hajipur, Vaishali 844102, India
| | - Ashish
- The Council of Scientific and Industrial Research Institute of Microbial Technology, Chandigarh, India
| | - Pradeep Das
- Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Bishwajit Kundu
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi 110016, India.
| |
Collapse
|
28
|
de Araujo ED, Manaswiyoungkul P, Israelian J, Park J, Yuen K, Farhangi S, Berger-Becvar A, Abu-Jazar L, Gunning PT. High-throughput thermofluor-based assays for inhibitor screening of STAT SH2 domains. J Pharm Biomed Anal 2017; 143:159-167. [DOI: 10.1016/j.jpba.2017.04.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/27/2017] [Accepted: 04/30/2017] [Indexed: 11/24/2022]
|
29
|
Watson MD, Monroe J, Raleigh DP. Size-Dependent Relationships between Protein Stability and Thermal Unfolding Temperature Have Important Implications for Analysis of Protein Energetics and High-Throughput Assays of Protein–Ligand Interactions. J Phys Chem B 2017; 122:5278-5285. [DOI: 10.1021/acs.jpcb.7b05684] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | - Daniel P. Raleigh
- Research Department of Structural and Molecular Biology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| |
Collapse
|
30
|
Deibler KK, Mishra RK, Clutter MR, Antanasijevic A, Bergan R, Caffrey M, Scheidt KA. A Chemical Probe Strategy for Interrogating Inhibitor Selectivity Across the MEK Kinase Family. ACS Chem Biol 2017; 12:1245-1256. [PMID: 28263556 PMCID: PMC5652073 DOI: 10.1021/acschembio.6b01060] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
MEK4 is an upstream kinase in MAPK signaling pathways where it phosphorylates p38 MAPK and JNK in response to mitogenic and cellular stress queues. MEK4 is overexpressed and induces metastasis in advanced prostate cancer lesions. However, the value of MEK4 as an oncology target has not been pharmacologically validated because selective chemical probes targeting MEK4 have not been developed. Despite a high level of sequence homology in the ATP-binding site, most reported MEK inhibitors are selective for MEK1/2 and display reduced potency toward other MEKs. Here, we present the first functional and binding selectivity-profiling platform of the MEK family. We applied the platform to profile a set of known kinase inhibitors and used the results to develop an in silico approach for small molecule docking against MEK proteins. The docking studies identified molecular features of the ligands and corresponding amino acids in MEK proteins responsible for high affinity binding versus those driving selectivity. WaterLOGSY and saturation transfer difference (STD) NMR spectroscopy techniques were utilized to understand the binding modes of active compounds. Further minor synthetic manipulations provide a proof of concept by showing how information gained through this platform can be utilized to perturb selectivity across the MEK family. This inhibitor-based approach pinpoints key features governing MEK family selectivity and clarifies empirical selectivity profiles for a set of kinase inhibitors. Going forward, the platform provides a rationale for facilitating the development of MEK-selective inhibitors, particularly MEK4 selective inhibitors, and repurposing of kinase inhibitors for probing the structural selectivity of isoforms.
Collapse
Affiliation(s)
- Kristine K. Deibler
- Department of Chemistry, Northwestern University, Evanston, 60208, Illinois, United States
| | - Rama K. Mishra
- Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, United States
| | - Matthew R. Clutter
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, United States
| | - Aleksandar Antanasijevic
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland Ave, Chicago, Illinois 60607, United States
| | - Raymond Bergan
- Knight Cancer Institute, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Rd., Portland, Oregon 97239, United States
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland Ave, Chicago, Illinois 60607, United States
| | - Karl A. Scheidt
- Department of Chemistry, Northwestern University, Evanston, 60208, Illinois, United States
- Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, United States
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, United States
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| |
Collapse
|
31
|
Icaritin enhances mESC self-renewal through upregulating core pluripotency transcription factors mediated by ERα. Sci Rep 2017; 7:40894. [PMID: 28091581 PMCID: PMC5238509 DOI: 10.1038/srep40894] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/12/2016] [Indexed: 12/28/2022] Open
Abstract
Utilization of small molecules in modulation of stem cell self-renewal is a promising approach to expand stem cells for regenerative therapy. Here, we identify Icaritin, a phytoestrogen molecule enhances self-renewal of mouse embryonic stem cells (mESCs). Icaritin increases mESCs proliferation while maintains their self-renewal capacity in vitro and pluripotency in vivo. This coincides with upregulation of key pluripotency transcription factors OCT4, NANOG, KLF4 and SOX2. The enhancement of mESCs self-renewal is characterized by increased population in S-phase of cell cycle, elevation of Cylin E and Cyclin-dependent kinase 2 (CDK2) and downregulation of p21, p27 and p57. PCR array screening reveals that caudal-related homeobox 2 (Cdx2) and Rbl2/p130 are remarkably suppressed in mESCs treated with Icaritin. siRNA knockdown of Cdx2 or Rbl2/p130 upregulates the expression of Cyclin E, OCT4 and SOX2, and subsequently increases cell proliferation and colony forming efficiency of mESCs. We then demonstrate that Icaritin co-localizes with estrogen receptor alpha (ERα) and activates its nuclear translocation in mESCs. The promotive effect of Icaritin on cell cycle and pluripotency regulators are eliminated by siRNA knockdown of ERα in mESCs. The results suggest that Icaritin enhances mESCs self-renewal by regulating cell cycle machinery and core pluripotency transcription factors mediated by ERα.
Collapse
|
32
|
Cleenewerck M, Grootaert MO, Gladysz R, Adriaenssens Y, Roelandt R, Joossens J, Lambeir AM, De Meyer GR, Declercq W, Augustyns K, Martinet W, Van der Veken P. Inhibitor screening and enzymatic activity determination for autophagy target Atg4B using a gel electrophoresis-based assay. Eur J Med Chem 2016; 123:631-638. [DOI: 10.1016/j.ejmech.2016.07.073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 07/15/2016] [Accepted: 07/28/2016] [Indexed: 12/19/2022]
|
33
|
Shrivastava P, Navratna V, Silla Y, Dewangan RP, Pramanik A, Chaudhary S, Rayasam G, Kumar A, Gopal B, Ramachandran S. Inhibition of Mycobacterium tuberculosis dihydrodipicolinate synthase by alpha-ketopimelic acid and its other structural analogues. Sci Rep 2016; 6:30827. [PMID: 27501775 PMCID: PMC4977564 DOI: 10.1038/srep30827] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/11/2016] [Indexed: 11/09/2022] Open
Abstract
The Mycobacterium tuberculosis dihydrodipicolinate synthase (Mtb-dapA) is an essential gene. Mtb-DapA catalyzes the aldol condensation between pyruvate and L-aspartate-beta-semialdehyde (ASA) to yield dihydrodipicolinate. In this work we tested the inhibitory effects of structural analogues of pyruvate on recombinant Mtb-DapA (Mtb-rDapA) using a coupled assay with recombinant dihydrodipicolinate reductase (Mtb-rDapB). Alpha-ketopimelic acid (α-KPA) showed maximum inhibition of 88% and IC50 of 21 μM in the presence of pyruvate (500 μM) and ASA (400 μM). Competition experiments with pyruvate and ASA revealed competition of α-KPA with pyruvate. Liquid chromatography-mass spectrometry (LC-MS) data with multiple reaction monitoring (MRM) showed that the relative abundance peak of final product, 2,3,4,5-tetrahydrodipicolinate, was decreased by 50%. Thermal shift assays showed 1 °C Tm shift of Mtb-rDapA upon binding α-KPA. The 2.4 Å crystal structure of Mtb-rDapA-α-KPA complex showed the interaction of critical residues at the active site with α-KPA. Molecular dynamics simulations over 500 ns of pyruvate docked to Mtb-DapA and of α-KPA-bound Mtb-rDapA revealed formation of hydrogen bonds with pyruvate throughout in contrast to α-KPA. Molecular descriptors analysis showed that ligands with polar surface area of 91.7 Å(2) are likely inhibitors. In summary, α-hydroxypimelic acid and other analogues could be explored further as inhibitors of Mtb-DapA.
Collapse
Affiliation(s)
- Priyanka Shrivastava
- Functional Genomics Unit, Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology (CSIR-IGIB), South Campus, New Delhi 110025, India
- Academy of Scientific and Innovative Research, CSIR-IGIB South Campus, New Delhi 110025, India
| | - Vikas Navratna
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Yumnam Silla
- Biotechnology Group (BIF center), Biological Science & Technology Division (BSTD), CSIR-North-East Institute of Science and Technology (CSIR-NEIST), Jorhat, Assam, 785006, India
| | - Rikeshwer P. Dewangan
- Functional Genomics Unit, Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology (CSIR-IGIB), South Campus, New Delhi 110025, India
| | - Atreyi Pramanik
- Functional Genomics Unit, Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology (CSIR-IGIB), South Campus, New Delhi 110025, India
| | - Sarika Chaudhary
- Functional Genomics Unit, Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology (CSIR-IGIB), South Campus, New Delhi 110025, India
| | - GeethaVani Rayasam
- Open Source Drug Discovery Unit (OSDD), CSIR-IGIB, New Delhi, 110001, India
| | - Anuradha Kumar
- Open Source Drug Discovery Unit (OSDD), CSIR-IGIB, New Delhi, 110001, India
| | | | - Srinivasan Ramachandran
- Functional Genomics Unit, Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology (CSIR-IGIB), South Campus, New Delhi 110025, India
- Academy of Scientific and Innovative Research, CSIR-IGIB South Campus, New Delhi 110025, India
| |
Collapse
|
34
|
Abstract
Protein phosphorylation lies at the heart of cell signalling, and somatic mutation(s) in kinases drives and sustains a multitude of human diseases, including cancer. The human protein kinase superfamily (the kinome) encodes approximately 50 'pseudokinases', which were initially predicted to be incapable of dynamic cell signalling when compared with canonical enzymatically active kinases. This assumption was supported by bioinformatics, which showed that amino acid changes at one or more key loci, making up the nucleotide-binding site or phosphotransferase machinery, were conserved in multiple vertebrate and non-vertebrate pseudokinase homologues. Protein kinases are highly attractive targets for drug discovery, as evidenced by the approval of almost 30 kinase inhibitors in oncology, and the successful development of the dual JAK1/2 (Janus kinase 1/2) inhibitor ruxolitinib for inflammatory indications. However, for such a large (>550) protein family, a remarkable number have still not been analysed at the molecular level, and only a surprisingly small percentage of kinases have been successfully targeted clinically. This is despite evidence that many are potential candidates for the development of new therapeutics. Indeed, several recent reports confirm that disease-associated pseudokinases can bind to nucleotide co-factors at concentrations achievable in the cell. Together, these findings suggest that drug targeting using either ATP-site or unbiased ligand-discovery approaches should now be attempted using the validation technology currently employed to evaluate their classic protein kinase counterparts. In the present review, we discuss members of the human pseudokinome repertoire, and catalogue somatic amino acid pseudokinase mutations that are emerging as the depth and clinical coverage of the human cancer pseudokinome expand.
Collapse
|
35
|
McKellar JLO, Minnell JJ, Gerth ML. A high‐throughput screen for ligand binding reveals the specificities of three amino acid chemoreceptors from
P
seudomonas syringae
pv.
actinidiae. Mol Microbiol 2015; 96:694-707. [DOI: 10.1111/mmi.12964] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2015] [Indexed: 11/30/2022]
Affiliation(s)
- James L. O. McKellar
- Department of Biochemistry University of Otago PO Box 56 Dunedin 9054 New Zealand
| | - Jordan J. Minnell
- Department of Biochemistry University of Otago PO Box 56 Dunedin 9054 New Zealand
| | - Monica L. Gerth
- Department of Biochemistry University of Otago PO Box 56 Dunedin 9054 New Zealand
| |
Collapse
|
36
|
Abstract
Targeting prostate cancer metastasis has very high therapeutic potential. Prostate cancer is the second most common cause of cancer death among men in the USA, and death results from the development of metastatic disease. In order to metastasize, cancer cells must complete a series of steps that together constitute the metastatic cascade. Each step therefore offers the opportunity for therapeutic targeting. However, practical limitations have served as limiting roadblocks to successfully targeting the metastatic cascade. They include our still-emerging understanding of the underlying biology, as well as the fact that many of the dysregulated processes have critical functionality in otherwise normal cells. We provide a discussion of the underlying biology, as it relates to therapeutic targeting. Therapeutic inroads are rapidly being made, and we present a series of case studies to highlight key points. Finally, future perspectives related to drug discovery for antimetastatic agents are discussed.
Collapse
|