1
|
Shekhar A, Di Lucrezia R, Jerye K, Korotkov VS, Harmrolfs K, Rox K, Weich HA, Ghai I, Delhommel F, Becher I, Degenhart C, Fansa E, Unger A, Habenberger P, Klebl B, Lukat P, Schmelz S, Henke S, Borgert S, Lang JC, Sasse F, Diestel R, Richter C, Schneider-Daum N, Hinkelmann B, Niemz J, Lehr CM, Jänsch L, Huehn J, Alm R, Savitski M, Welte T, Hesterkamp T, Sattler M, Winterhalter M, Blankenfeldt W, Medina E, Bilitewski U, Dinkel K, Brönstrup M. Highly potent quinoxalinediones inhibit α-hemolysin and ameliorate Staphylococcus aureus lung infections. Cell Host Microbe 2025; 33:560-572.e21. [PMID: 40168998 DOI: 10.1016/j.chom.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/27/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025]
Abstract
Hospital-acquired pneumonia caused by Staphylococcus aureus is associated with patient morbidity and mortality, despite adequate antibiotic therapy. This illustrates the need for treatments beyond antibiotics. The pore-forming heptameric toxin α-hemolysin (Hla) is a major pathogenicity factor of S. aureus and a clinically validated target. We identify quinoxalinediones (QDS) as highly potent Hla inhibitors, conferring protection against the hallmarks of Hla-induced pathogenicity such as Ca2+ influx, cytotoxicity, hemolysis, and monolayer destruction. The effects were exerted across major Hla subtypes in all relevant cell types. QDS prevented the formation of functional pores by interacting with Hla near the phospholipid-binding site. The QDS analog, H052, was active in mouse models of S. aureus lung infections, when administered prophylactically or therapeutically, either as monotherapy or when given in combination with the antibiotic linezolid. The study provides evidence that complex bacterial toxins can be targeted in vivo by drug-like small molecules.
Collapse
Affiliation(s)
- Aditya Shekhar
- Compound Profiling and Screening, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | | | - Karoline Jerye
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Vadim S Korotkov
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Kirsten Harmrolfs
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Katharina Rox
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany; German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Braunschweig, Germany
| | - Herbert A Weich
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Ishan Ghai
- Life Sciences and Chemistry, Constructor University, 28759 Bremen, Germany
| | - Florent Delhommel
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Bavarian NMR Center, Lichtenbergstrasse 4, 85747 Garching, Germany; Helmholtz Munich, Molecular Targets and Therapeutics Center, Institute of Structural Biology, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | - Isabelle Becher
- Proteomics Core Facility, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | | | - Eyad Fansa
- Lead Discovery Center, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Anke Unger
- Lead Discovery Center, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | | | - Bert Klebl
- Lead Discovery Center, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Peer Lukat
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Stefan Schmelz
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Steffi Henke
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Sebastian Borgert
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Julia C Lang
- Infection Immunology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Florenz Sasse
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Randi Diestel
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Clémentine Richter
- Department of Drug Delivery, Helmholtz-Institute for Pharmaceutical Research (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Nicole Schneider-Daum
- Department of Drug Delivery, Helmholtz-Institute for Pharmaceutical Research (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Bettina Hinkelmann
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Jana Niemz
- Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz-Institute for Pharmaceutical Research (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Lothar Jänsch
- Cellular Proteomics, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany; Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | | | - Mikhail Savitski
- Proteomics Core Facility, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Tobias Welte
- Department of Respiratory Medicine and Infectious Disease, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover 30625, Germany
| | - Thomas Hesterkamp
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Braunschweig, Germany
| | - Michael Sattler
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Bavarian NMR Center, Lichtenbergstrasse 4, 85747 Garching, Germany; Helmholtz Munich, Molecular Targets and Therapeutics Center, Institute of Structural Biology, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | | | - Wulf Blankenfeldt
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Eva Medina
- Infection Immunology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Ursula Bilitewski
- Compound Profiling and Screening, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Klaus Dinkel
- Lead Discovery Center, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Mark Brönstrup
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany; German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Braunschweig, Germany; Center of Biomolecular Drug Research (BMWZ), Leibniz Universität, 30167 Hannover, Germany.
| |
Collapse
|
2
|
Nettey-Oppong EE, Muhammad R, Ali A, Jeong HW, Seok YS, Kim SW, Choi SH. The Impact of Temperature and Pressure on the Structural Stability of Solvated Solid-State Conformations of Bombyx mori Silk Fibroins: Insights from Molecular Dynamics Simulations. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5686. [PMID: 39685120 DOI: 10.3390/ma17235686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024]
Abstract
Bombyx mori silk fibroin is a promising biopolymer with notable mechanical strength, biocompatibility, and potential for diverse biomedical applications, such as tissue engineering scaffolds, and drug delivery. These properties are intrinsically linked to the structural characteristics of silk fibroin, making it essential to understand its molecular stability under varying environmental conditions. This study employed molecular dynamics simulations to examine the structural stability of silk I and silk II conformations of silk fibroin under changes in temperature (298 K to 378 K) and pressure (0.1 MPa to 700 MPa). Key parameters, including Root Mean Square Deviation (RMSD), Root Mean Square Fluctuation (RMSF), and Radius of Gyration (Rg) were analyzed, along with non-bonded interactions such as van der Waals and electrostatic potential energy. Our findings demonstrate that both temperature and pressure exert a destabilizing effect on silk fibroin, with silk I exhibiting a higher susceptibility to destabilization compared to silk II. Additionally, pressure elevated the van der Waals energy in silk I, while temperature led to a reduction. In contrast, electrostatic potential energy remained unaffected by these environmental conditions, highlighting stable long-range interactions throughout the study. Silk II's tightly packed β-sheet structure offers greater resilience to environmental changes, while the more flexible α-helices in silk I make it more susceptible to structural perturbations. These findings provide valuable insights into the atomic-level behavior of silk fibroin, contributing to a deeper understanding of its potential for applications in environments where mechanical or thermal stress is a factor. The study underscores the importance of computational approaches in exploring protein stability and supports the continued development of silk fibroin for biomedical and engineering applications.
Collapse
Affiliation(s)
| | - Riaz Muhammad
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Ahmed Ali
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
- Department of Electrical Engineering, Sukkur IBA University, Sukkur 65200, Pakistan
| | - Hyun-Woo Jeong
- Department of Biomedical Engineering, Eulji University, Seongnam 13135, Republic of Korea
| | - Young-Seek Seok
- Gangwon-do Agricultural Product Registered Seed Station, Chuncheon 24410, Republic of Korea
| | - Seong-Wan Kim
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Republic of Korea
| | - Seung Ho Choi
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
- Department of Integrative Medicine, Major in Digital Healthcare, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| |
Collapse
|
3
|
Vadakkan K, Sathishkumar K, Kuttiyachan Urumbil S, Ponnenkunnathu Govindankutty S, Kumar Ngangbam A, Devi Nongmaithem B. A review of chemical signaling mechanisms underlying quorum sensing and its inhibition in Staphylococcus aureus. Bioorg Chem 2024; 148:107465. [PMID: 38761705 DOI: 10.1016/j.bioorg.2024.107465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/29/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
Staphylococcus aureus is a significant bacterium responsible for multiple infections and is a primary cause of fatalities among patients in hospital environments. The advent of pathogenic bacteria such as methicillin-resistant S. aureus revealed the shortcomings of employing antibiotics to treat bacterial infectious diseases. Quorum sensing enhances S. aureus's survivability through signaling processes. Targeting the key components of quorum sensing has drawn much interest nowadays as a promising strategy for combating infections caused by bacteria. Concentrating on the accessory gene regulator quorum-sensing mechanism is the most commonly suggested anti-virulence approach for S.aureus. Quorum quenching is a common strategy for controlling illnesses triggered by microorganisms since it reduces the pathogenicity of bacteria and improves bacterial biofilm susceptibility to antibiotics, thus providing an intriguing prospect for drug discovery. Quorum sensing inhibition reduces selective stresses and constrains the emergence of antibiotic resistance while limiting bacterial pathogenicity. This review examines the quorum sensing mechanisms involved in S. aureus, quorum sensing targets and gene regulation, environmental factors affecting quorum sensing, quorum sensing inhibition, natural products as quorum sensing inhibitory agents and novel therapeutical strategies to target quorum sensing in S. aureus as drug developing technique to augment conventional antibiotic approaches.
Collapse
Affiliation(s)
- Kayeen Vadakkan
- Department of Biotechnology, St. Mary's College (Autonomous), Thrissur, Kerala 680020, India; Manipur International University, Imphal, Manipur 795140, India.
| | - Kuppusamy Sathishkumar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Thandalam, Chennai, Tamil Nadu 602105, India
| | | | | | | | | |
Collapse
|
4
|
Paternoster C, Tarenzi T, Potestio R, Lattanzi G. Gamma-Hemolysin Components: Computational Strategies for LukF-Hlg2 Dimer Reconstruction on a Model Membrane. Int J Mol Sci 2023; 24:ijms24087113. [PMID: 37108277 PMCID: PMC10138441 DOI: 10.3390/ijms24087113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/07/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The gamma-hemolysin protein is one of the most common pore-forming toxins expressed by the pathogenic bacterium Staphylococcus aureus. The toxin is used by the pathogen to escape the immune system of the host organism, by assembling into octameric transmembrane pores on the surface of the target immune cell and leading to its death by leakage or apoptosis. Despite the high potential risks associated with Staphylococcus aureus infections and the urgent need for new treatments, several aspects of the pore-formation process from gamma-hemolysin are still unclear. These include the identification of the interactions between the individual monomers that lead to the formation of a dimer on the cell membrane, which represents the unit for further oligomerization. Here, we employed a combination of all-atom explicit solvent molecular dynamics simulations and protein-protein docking to determine the stabilizing contacts that guide the formation of a functional dimer. The simulations and the molecular modeling reveal the importance of the flexibility of specific protein domains, in particular the N-terminus, to drive the formation of the correct dimerization interface through functional contacts between the monomers. The results obtained are compared with the experimental data available in the literature.
Collapse
Affiliation(s)
- Costanza Paternoster
- Department of Physics, University of Trento, Via Sommarive 14, I-38123 Trento, Italy
- INFN-TIFPA, Trento Institute for Fundamental Physics and Applications, Via Sommarive 14, I-38123 Trento, Italy
| | - Thomas Tarenzi
- Department of Physics, University of Trento, Via Sommarive 14, I-38123 Trento, Italy
- INFN-TIFPA, Trento Institute for Fundamental Physics and Applications, Via Sommarive 14, I-38123 Trento, Italy
| | - Raffaello Potestio
- Department of Physics, University of Trento, Via Sommarive 14, I-38123 Trento, Italy
- INFN-TIFPA, Trento Institute for Fundamental Physics and Applications, Via Sommarive 14, I-38123 Trento, Italy
| | - Gianluca Lattanzi
- Department of Physics, University of Trento, Via Sommarive 14, I-38123 Trento, Italy
- INFN-TIFPA, Trento Institute for Fundamental Physics and Applications, Via Sommarive 14, I-38123 Trento, Italy
| |
Collapse
|
5
|
Electrophysiological and spectroscopic investigation of hydrolysable tannins interaction with α-hemolysin of S. aureus. Bioelectrochemistry 2023; 150:108318. [PMID: 36470005 DOI: 10.1016/j.bioelechem.2022.108318] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/04/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
In this study, using bilayer lipid membrane technique, we report a novel facet of antihemolytic activity of two tannins (1,2,3,4,5-penta-O-galloyl-β-D-glucose (PGG) and 1,2-di-O-galloyl-4,6-valoneoyl-β-D-glucose (dGVG)), which consists in inhibiting the formation of α-hemolysin channels and blocking the conductivity of already formed channels. These effects were observed at tannin concentrations well below minimal inhibitory concentration values for S. aureus growth. Using spectroscopic methods, we show that these two tannins differing in molecular structure but having the same number of -OH groups and aromatic rings form firm complexes with hemolysin in aqueous solutions, which may underlie the disruption of its subsequent interaction with the membrane, thus preventing hemolysis of erythrocytes. In all experimental settings, PGG was the more active compound compared to dGVG, that indicates the important role of the flexibility of the tannin molecule in interaction with the toxin. In addition, we found that PGG, but not dGVG, was able to block the release of the toxin by bacterial cells. This toxin is a strong pathogenic factor causing a number of diseases and therefore is considered as a virulence target for treatment of S. aureus infection, so the data obtained suggest that PGG and possibly other tannins of similar structure have therapeutic potential in fighting the virulence of S. aureus.
Collapse
|
6
|
Sabino YNV, Cotter PD, Mantovani HC. Anti-virulence compounds against Staphylococcus aureus associated with bovine mastitis: A new therapeutic option? Microbiol Res 2023; 271:127345. [PMID: 36889204 DOI: 10.1016/j.micres.2023.127345] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023]
Abstract
Bovine mastitis represents a major economic burden faced by the dairy industry. S. aureus is an important and prevalent bovine mastitis-associated pathogen in dairy farms worldwide. The pathogenicity and persistence of S. aureus in the bovine mammary gland are associated with the expression of a range of virulence factors involved in biofilm formation and the production of several toxins. The traditional therapeutic approach to treating bovine mastitis includes the use of antibiotics, but the emergence of antibiotic-resistant strains has caused therapeutic failure. New therapeutic approaches targeting virulence factors of S. aureus rather than cell viability can have several advantages including lower selective pressure towards the development of resistance and little impact on the host commensal microbiota. This review summarizes the potential of anti-virulence therapies to control S. aureus associated with bovine mastitis focusing on anti-toxin, anti-biofilm, and anti-quorum sensing compounds. It also points to potential sources of new anti-virulence inhibitors and presents screening strategies for identifying these compounds.
Collapse
Affiliation(s)
| | | | - Hilario C Mantovani
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
7
|
Sajeev A, Hegde M, Girisa S, Devanarayanan TN, Alqahtani MS, Abbas M, Sil SK, Sethi G, Chen JT, Kunnumakkara AB. Oroxylin A: A Promising Flavonoid for Prevention and Treatment of Chronic Diseases. Biomolecules 2022; 12:1185. [PMID: 36139025 PMCID: PMC9496116 DOI: 10.3390/biom12091185] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
There have been magnificent advancements in the understanding of molecular mechanisms of chronic diseases over the past several years, but these diseases continue to be a considerable cause of death worldwide. Most of the approved medications available for the prevention and treatment of these diseases target only a single gene/protein/pathway and are known to cause severe side effects and are less effective than they are anticipated. Consequently, the development of finer therapeutics that outshine the existing ones is far-reaching. Natural compounds have enormous applications in curbing several disastrous and fatal diseases. Oroxylin A (OA) is a flavonoid obtained from the plants Oroxylum indicum, Scutellaria baicalensis, and S. lateriflora, which have distinctive pharmacological properties. OA modulates the important signaling pathways, including NF-κB, MAPK, ERK1/2, Wnt/β-catenin, PTEN/PI3K/Akt, and signaling molecules, such as TNF-α, TGF-β, MMPs, VEGF, interleukins, Bcl-2, caspases, HIF-1α, EMT proteins, Nrf-2, etc., which play a pivotal role in the molecular mechanism of chronic diseases. Overwhelming pieces of evidence expound on the anti-inflammatory, anti-bacterial, anti-viral, and anti-cancer potentials of this flavonoid, which makes it an engrossing compound for research. Numerous preclinical and clinical studies also displayed the promising potential of OA against cancer, cardiovascular diseases, inflammation, neurological disorders, rheumatoid arthritis, osteoarthritis, etc. Therefore, the current review focuses on delineating the role of OA in combating different chronic diseases and highlighting the intrinsic molecular mechanisms of its action.
Collapse
Affiliation(s)
- Anjana Sajeev
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Thulasidharan Nair Devanarayanan
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
- BioImaging Unit, Space Research Center, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
- Electronics and Communications Department, College of Engineering, Delta University for Science and Technology, Gamasa 35712, Egypt
| | - Samir Kumar Sil
- Cell Physiology and Cancer Biology Laboratory, Department of Human Physiology, Tripura University, Suryamaninagar 799022, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Jen-Tsung Chen
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung 811, Taiwan
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| |
Collapse
|
8
|
Liu RP, Wang XQ, Wang J, Dan L, Li YH, Jiang H, Xu YN, Kim NH. Oroxin A reduces oxidative stress, apoptosis, and autophagy and improves the developmental competence of porcine embryos in vitro. Reprod Domest Anim 2022; 57:1255-1266. [PMID: 35780288 DOI: 10.1111/rda.14200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/01/2022] [Indexed: 11/28/2022]
Abstract
Oroxin A (OA) is a flavonoid isolated from Oroxylum indicum (L.) Kurz that has various biological activities, including antioxidant activities. This study aimed to examine the viability of using OA in an in vitro culture (IVC) medium for its antioxidant effects and related molecular mechanisms on porcine blastocyst development. In this study, we investigated the effects of OA on early porcine embryo development via terminal deoxynucleotidyl transferase dUTP nick-end labeling, 5-ethynyl-2'-deoxyuridine labeling, quantitative reverse transcription PCR, and immunocytochemistry. Embryos cultured in the IVC medium supplemented with 2.5 μM of OA had an increased blastocyst formation rate, total cell number, and proliferation capacity, along with a low apoptosis rate. OA supplementation decreased reactive oxygen species levels, while increasing glutathione levels. OA-treated embryos exhibited an improved intracellular mitochondrial membrane potential and reduced autophagy. Moreover, levels of pluripotency- and antioxidant-related genes were upregulated, whereas those of apoptosis- and autophagy-related genes were downregulated by OA addition. In conclusion, OA improves preimplantation embryonic development by reducing oxidative stress and enhancing mitochondrial function.
Collapse
Affiliation(s)
- Rong-Ping Liu
- School of Biotechnology and Health Sciences, Wuyi University, 529000, Jiangmen, China
| | - Xin-Qin Wang
- School of Biotechnology and Health Sciences, Wuyi University, 529000, Jiangmen, China
| | - Jing Wang
- School of Biotechnology and Health Sciences, Wuyi University, 529000, Jiangmen, China
| | - Luo Dan
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, China
| | - Ying-Hua Li
- School of Biotechnology and Health Sciences, Wuyi University, 529000, Jiangmen, China
| | - Hao Jiang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, China
| | - Yong-Nan Xu
- School of Biotechnology and Health Sciences, Wuyi University, 529000, Jiangmen, China
| | - Nam-Hyung Kim
- School of Biotechnology and Health Sciences, Wuyi University, 529000, Jiangmen, China
| |
Collapse
|
9
|
Cheng X, Huang T, Wang C, Hao S, Shu L, Wang S, Cheng G, Zhang Q, Huang J, Chen C. Natural Compound Library Screening Identifies Oroxin A for the Treatment of Myocardial Ischemia/Reperfusion Injury. Front Pharmacol 2022; 13:894899. [PMID: 35645816 PMCID: PMC9133817 DOI: 10.3389/fphar.2022.894899] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/19/2022] [Indexed: 12/20/2022] Open
Abstract
Myocardial ischemia/reperfusion injury (MI/RI) is a serious pathophysiological process relating to cardiovascular disease. Oroxin A (OA) is a natural flavonoid glycoside with various biological activities. However, its effect on the pathophysiological process of MI/RI has not yet been reported. The aim of this study was to determine whether OA could alleviate MI/RI induced inflammation and pyroptosis in vivo and in vitro, providing a novel therapeutic regimen for the treatment of MI/RI. A high-throughput drug screening strategy was employed to test 2,661 natural compound libraries that can alleviate MI/RI in vivo and in vitro. The rat model of MI/RI was established by ligating the left anterior descending (LAD) coronary artery. H9c2 cells were subjected to oxygen-glucose deprivation/reperfusion (OGD/R) to simulate MI/RI. The results show that OA is able to significantly inhibit apoptosis, pyroptosis and the inflammation response (TNF-α, IL-6, IL-8, IL-10, IL-1β, IL-18) in vivo and in vitro, and reduce the release of myocardial enzymes (cTnI, cTnT, CK-MB, LDH, AST). In the rat MI/RI model, OA can not only improve cardiac function and reduce inflammatory cell infiltration but also reduce myocardial infarct size. The results revealed that OA is an effective remedy against MI/RI as it reduces the inflammatory response and inhibits pyroptosis. This may provide a new therapeutic target for the clinical treatment of MI/RI.
Collapse
Affiliation(s)
- Xingdong Cheng
- Department of Anesthesiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Tingting Huang
- Department of Anesthesiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chunhui Wang
- Department of Anesthesiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shuang Hao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liliang Shu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shixiong Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Gao Cheng
- Department of Anesthesiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiaoyun Zhang
- Department of Anesthesiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jian Huang
- Department of Anesthesiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
- *Correspondence: Chen Chen, ; Jian Huang,
| | - Chen Chen
- Department of Anesthesiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Chen Chen, ; Jian Huang,
| |
Collapse
|
10
|
Huang JM, Wang CZ, Lu SY, Wang Z, Yan ZQ. Oroxin B Attenuates Ovariectomy-Induced Bone Loss by Suppressing Osteoclast Formation and Activity. Drug Des Devel Ther 2021; 15:4811-4825. [PMID: 34876805 PMCID: PMC8643139 DOI: 10.2147/dddt.s328238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/16/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Osteoclasts are the major players in bone resorption and have always been studied in the prevention and treatment of osteoporosis. Previous studies have confirmed that a variety of flavonoids inhibit osteoporosis and improve bone health mainly through inhibiting osteoclastogenesis. Oroxin B (OB) is a flavonoid compound extracted from traditional Chinese herbal medicine Oroxylum indicum (L.) Vent, exerts potent antitumor and anti-inflammation effect, but its effect on osteoclastogensis remains unknown. METHODS We comprehensively evaluated the effect of OB on the formation and function of osteoclasts and the underling mechanism by bone marrow-derived macrophage in vitro. In vivo, we used mice ovariectomized model to verify the protective effect of OB. RESULTS OB was found to inhibit osteoclast formation and bone resorption function in vitro, in a dose-dependent manner and the increased osteoclastic-related genes induced by RANKL (NFATc1, c-fos, cathepsin K, RANK, MMP9 and TRAP) were also attenuated following OB treatment. Mechanistical investigation showed OB abrogated the increased phosphorylation level of MAPK and NF-κB pathway, and diminished the expression of the vital transcription factors for osteoclastogenesis. OB also prevented ovariectomy (OVX)-induced bone loss by inhibiting osteoclast formation and activity in mice. CONCLUSION Our study demonstrated that OB may act as an anti-osteoporosis agent by inhibiting osteoclast maturation and attenuating bone resorption.
Collapse
Affiliation(s)
- Jun-ming Huang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Chen-zhong Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Shun-yi Lu
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Zhe Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Zuo-qin Yan
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
11
|
Sannigrahi A, Chattopadhyay K. Pore formation by pore forming membrane proteins towards infections. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:79-111. [PMID: 35034727 DOI: 10.1016/bs.apcsb.2021.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Over the last 25 years, the biology of membrane proteins, including the PFPs-membranes interactions is seeking attention for the development of successful drug molecules against a number of infectious diseases. Pore forming toxins (PFTs), the largest family of PFPs are considered as a group of virulence factors produced in a large number of pathogenic systems which include streptococcus, pneumonia, Staphylococcus aureus, E. coli, Mycobacterium tuberculosis, group A and B streptococci, Corynebacterium diphtheria and many more. PFTs are generally utilized by the disease causing pathogens to disrupt the host first line of defense i.e. host cell membranes through pore formation strategy. Although, pore formation is the principal mode of action of the PFTs but they can have additional adverse effects on the hosts including immune evasion. Recently, structural investigation of different PFTs have imparted the molecular mechanistic insights into how PFTs get transformed from its inactive state to active toxic state. On the basis of their structural entity, PFTs have been classified in different types and their mode of actions alters in terms of pore formation and corresponding cellular toxicity. Although pathogen genome analysis can identify the probable PFTs depending upon their structural diversity, there are so many PFTs which utilize the local environmental conditions to generate their pore forming ability using a novel strategy which is known as "conformational switch" of a protein. This conformational switch is considered as characteristics of the phase shifting proteins which were often utilized by many pathogenic systems to protect them from the invaders through allosteric communication between distant regions of the protein. In this chapter, we discuss the structure function relationships of PFTs and how activity of PFTs varies with the change in the environmental conditions has been explored. Finally, we demonstrate these structural insights to develop therapeutic potential to treat the infections caused by multidrug resistant pathogens.
Collapse
Affiliation(s)
- Achinta Sannigrahi
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, Karnataka, India.
| | - Krishnananda Chattopadhyay
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India.
| |
Collapse
|
12
|
Ahmad-Mansour N, Loubet P, Pouget C, Dunyach-Remy C, Sotto A, Lavigne JP, Molle V. Staphylococcus aureus Toxins: An Update on Their Pathogenic Properties and Potential Treatments. Toxins (Basel) 2021; 13:677. [PMID: 34678970 PMCID: PMC8540901 DOI: 10.3390/toxins13100677] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 01/12/2023] Open
Abstract
Staphylococcus aureus is a clinically important pathogen that causes a wide range of human infections, from minor skin infections to severe tissue infection and sepsis. S. aureus has a high level of antibiotic resistance and is a common cause of infections in hospitals and the community. The rising prevalence of community-acquired methicillin-resistant S. aureus (CA-MRSA), combined with the important severity of S. aureus infections in general, has resulted in the frequent use of anti-staphylococcal antibiotics, leading to increasing resistance rates. Antibiotic-resistant S. aureus continues to be a major health concern, necessitating the development of novel therapeutic strategies. S. aureus uses a wide range of virulence factors, such as toxins, to develop an infection in the host. Recently, anti-virulence treatments that directly or indirectly neutralize S. aureus toxins have showed promise. In this review, we provide an update on toxin pathogenic characteristics, as well as anti-toxin therapeutical strategies.
Collapse
Affiliation(s)
- Nour Ahmad-Mansour
- Laboratory of Pathogen Host Interactions, CNRS UMR5235, Université de Montpellier, 34000 Montpellier, France;
| | - Paul Loubet
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Infectious and Tropical Diseases, Université de Montpellier, 30908 Nîmes, France; (P.L.); (A.S.)
| | - Cassandra Pouget
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Université de Montpellier, 30908 Nîmes, France;
| | - Catherine Dunyach-Remy
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Microbiology and Hospital Hygiene, Université de Montpellier, 30908 Nîmes, France; (C.D.-R.); (J.-P.L.)
| | - Albert Sotto
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Infectious and Tropical Diseases, Université de Montpellier, 30908 Nîmes, France; (P.L.); (A.S.)
| | - Jean-Philippe Lavigne
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Microbiology and Hospital Hygiene, Université de Montpellier, 30908 Nîmes, France; (C.D.-R.); (J.-P.L.)
| | - Virginie Molle
- Laboratory of Pathogen Host Interactions, CNRS UMR5235, Université de Montpellier, 34000 Montpellier, France;
| |
Collapse
|
13
|
Hu H, Liu M, Sun S. Pore-Forming Toxins During Bacterial Infection: Molecular Mechanisms and Potential Therapeutic Targets. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3773-3781. [PMID: 34522083 PMCID: PMC8434828 DOI: 10.2147/dddt.s322393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022]
Abstract
Bacterial infections are predominantly treated with antibiotics, and resistance to antibiotics is becoming an increasing threat to our health. Pore-forming toxins (PFTs) are virulence factors secreted by many pathogenic bacterial strains, both in acute and chronic infections. They are special membrane-targeting proteins that exert toxic effects by forming pores in the cell membrane. Recent studies have elucidated the structure of PFTs and the detailed molecular mechanisms of their pathogenicity. Here, we discuss recent findings that highlight the regulatory mechanisms and important roles of two types of PFTs, α-PFTs and β-PFTs, in mediating the virulence of bacteria, and the therapeutic potential of targeting PFTs for antibacterial treatment. Therapeutic strategies based on PFTs are highly specific and may alleviate the issue of increasing resistance to antibiotics.
Collapse
Affiliation(s)
- Haijie Hu
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - Min Liu
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - Shuang Sun
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, People's Republic of China
| |
Collapse
|
14
|
Algburi A, Al-Hasani HM, Ismael TK, Abdelhameed A, Weeks R, Ermakov AM, Chikindas ML. Antimicrobial Activity of Bacillus subtilis KATMIRA1933 and Bacillus amyloliquefaciens B-1895 Against Staphylococcus aureus Biofilms Isolated from Wound Infection. Probiotics Antimicrob Proteins 2021; 13:125-134. [PMID: 32556931 DOI: 10.1007/s12602-020-09673-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Staphylococcal wound infections range from mild to severe with life-threatening complications. The challenge of controlling such infections is related to bacterial biofilm formation, which is a major factor contributing to antibiotic resistance and infection recurrence. In this study, four clinical isolates of staphylococci species; two isolates of methicillin-resistant Staphylococcus aureus (MRSA) and two methicillin-sensitive Staphylococcus aureus (MSSA) isolates. The identification of bacterial species based on cell morphology, initial biochemical tests, and the VITEK2 system were used to confirm the clinical microbiological diagnosis. Antibiotic sensitivity testing showed that the isolated staphylococci were highly resistant to the following antibiotics, amoxicillin, penicillin G, cefotaxime, and methicillin. Combinations of cefotaxime with the cell-free supernatants (CFS) of Bacillus subtilis KATMIRA1933 and Bacillus amyloliquefaciens B-1895, each one separately showed complementary activity against the tested staphylococci. The co-aggregation capability of the tested bacilli as beneficial bacteria against isolated staphylococci was also evaluated. The data showed a strong co-aggregation with scores (+ 3, + 4) which were reported between the bacilli strains and the isolated staphylococci. Furthermore, the CFS of bacilli strains showed an inhibitory effect against biofilm-associated MRSA and MSSA. These findings confirmed the ability of beneficial bacteria to compete with the pathogens at the site of colonization or for the source of nutrients and, eventually, lead to inhibition of the pathogens' capability of causing a wound infection. Such beneficial bacteria could play an important role in future pharmaceutical and industrial applications.
Collapse
Affiliation(s)
- Ammar Algburi
- Department of Biotechnology, College of Science, University of Diyala, Baqubah, Iraq. .,Department of Scholarship and Cultural Relations, Presidency of Diyala University, Baqubah, Iraq.
| | - Halah M Al-Hasani
- Department of Biotechnology, College of Science, University of Diyala, Baqubah, Iraq
| | - Thurya K Ismael
- Educational Laboratories, General Teaching Hospital of Baqubah, Baqubah, Iraq
| | - Alyaa Abdelhameed
- Department of Biotechnology, College of Science, University of Diyala, Baqubah, Iraq
| | - Richard Weeks
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA
| | | | - Michael L Chikindas
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA.,Don State Technical University, Rostov-on-Don, Russia
| |
Collapse
|
15
|
Mohan R, Venugopal S. Molecular Binding and Simulation Studies of Staphylococcus aureus Superantigens with Flavonoid Compounds. Infect Disord Drug Targets 2021; 20:531-542. [PMID: 30727923 DOI: 10.2174/1871526519666190207092307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/02/2019] [Accepted: 02/01/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Superantigens of Staphylococcus aureus namely enterotoxin A, exfoliative toxin A, and Toxic shock syndrome toxin-1 cause detrimental effects on the cells of the immune system. METHODS In this work, the toxins were downloaded from the Protein DataBank database and energies were minimized using KoBaMIN server. Forty flavonoids compounds were identified by pubchem compound database through extensive literature study and their 3D structures were obtained by submitting SMILES to CORINA tool. Based on Lipinski's rule of five, the molecules were filtered that resulted in 27 compounds. Molecular docking was performed for identifying the binding and interaction sites of flavonoids with the toxins using Autodock 4. RESULTS AND CONCLUSION The docked complexes were then subjected to molecular dynamics simulation using Gromacs. The analysis revealed the stability of the complexes as indicated by three hydrogen bonds formed during the simulation time period of 20 ns.
Collapse
Affiliation(s)
- Ramadevi Mohan
- Department of Integrative Biology, School of Biosciences and Technology, VIT, Vellore, Tamil nadu, 632 014, India
| | - Subhashree Venugopal
- Department of Integrative Biology, School of Biosciences and Technology, VIT, Vellore, Tamil nadu, 632 014, India
| |
Collapse
|
16
|
Sithisarn P, Rojsanga P, Sithisarn P. Flavone-Rich Fractions and Extracts from Oroxylum indicum and Their Antibacterial Activities against Clinically Isolated Zoonotic Bacteria and Free Radical Scavenging Effects. Molecules 2021; 26:1773. [PMID: 33809943 PMCID: PMC8004265 DOI: 10.3390/molecules26061773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 12/01/2022] Open
Abstract
Oroxylum indicum extracts from the seeds collected from Lampang and Pattani provinces in Thailand, and young fruits and flowers exhibited in vitro display antioxidant and antibacterial activities against clinically isolated zoonotic bacteria including Staphylococcus intermedius, Streptococcus suis, Pseudomonas aeruginosa, β-hemolytic Escherichia coli and Staphylococcus aureus. The orange crystals and yellow precipitates were obtained from the preparation processes of the seed extracts. The orange-red crystals from the seeds collected from Lampang province exhibited strong in vitro 2,2-diphenyl-1-picrylhydrazyl (DPPH) scavenging effects (EC50 value = 25.99 ± 3.30 μg/mL) and antibacterial effects on S. intermedius and β-hemolytic E. coli while the yellow precipitate from the same source exhibited only antioxidant activity. Quantitative analysis of phytochemicals in O. indicum samples by spectrophotometric and HPLC techniques showed that they contained different amounts of total phenolic, total flavonoid and three major flavones; baicalin, baicalein and chrysin contents. Young fruit extract, which contained low amounts of flavone contents, still promoted antibacterial effects against the tested bacteria with IC50 values lower than 1 mg/mL and MIC values between 4 to 10 mg/mL in S. intermedius, S. aureus and S suis while higher IC50 and MIC values against P. aeruginosa and β-hemolytic E. coli were found. From scanning electron microscopy, the extract of the young fruit of O. indicum promoted morphological changes in the bacterial cells by disrupting the bacterial cell walls, inducing leakage of the cellular content, and generating the abnormal accumulation of cells. The mechanism of action of the extract for this antibacterial effect may be the disruption of the cell membrane and abnormal cell aggregations. Regression analysis of the results suggests the correlation between total phenolic and total flavonoid contents and antioxidant and antibacterial effects. Baicalin was found to have a high correlation with an inhibitory effect against β-hemolytic E. coli while three unidentified peaks, which could be flavones, showed high correlations with an inhibitory effect against S. intermedius, S. suis, P. aeruginosa and S. aureus.
Collapse
Affiliation(s)
- Patchima Sithisarn
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand;
| | - Piyanuch Rojsanga
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand;
| | - Pongtip Sithisarn
- Department of Pharmacognosy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
17
|
Wang G, Gao Y, Xu X, Zhang P, Wang J, Li G, Lv Q, Niu X, Liu H. Mode of action and structural modelling of the interaction of formononetin with suilysin. J Appl Microbiol 2021; 131:2010-2018. [PMID: 33639036 DOI: 10.1111/jam.15051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/02/2021] [Accepted: 02/17/2021] [Indexed: 11/28/2022]
Abstract
AIMS Suilysin is a critical pore-forming virulence factor of Streptococcus suis that has been demonstrated to substantially contribute to its pathogenicity. We have demonstrated that formononetin alleviates S. suis infection both in vivo and in vitro by targeting suilysin. However, the molecular mechanism of the effect is unclear. Our aim was to determine the molecular mechanism of the effect of formononetin on suilysin. METHODS AND RESULTS The mechanism of interaction between formononetin and suilysin was investigated by molecular modelling. The results indicated that formononetin was bound at the junction of domain two and domain four of suilysin. The binding free energy values indicated that the A415, Y412, E414, N413, T61, T62 and G416 residues are critical for this binding, this observation was confirmed by the changes in the flexibility of these residues and the distances between these residues and formononetin. The inhibitory effect of formononetin on the pore-forming activity of suilysin, binding constant and binding free energy were significantly decreased by site-specific mutagenesis of Y412 and N413. Finally, we analysed the spatial configuration of suilysin before and after formononetin binding, the results indicated that the binding changed the conformation of suilysin, especially the angle between domain two and domain four, resulting in the disruption of cholesterol binding to suilysin and in the loss of pore-forming activity. CONCLUSIONS Formononetin is located at the junction of domain two and domain four of suilysin, and Y412 and N413 play critical roles in the binding. Formononetin binding changes the angle between domain two and domain four of suilysin, resulting in the loss of the pore-inducing activity of suilysin. SIGNIFICANCE AND IMPACT OF THE STUDY This work will promote the application of formononetin to combat S. suis infections and may contribute to the development of new inhibitors or modification of existing inhibitors.
Collapse
Affiliation(s)
- G Wang
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, PR China.,College of Food Engineering, Jilin Engineering Normal University, Changchun, China.,College of Veterinary Medicine, Jilin University, Changchun, China
| | - Y Gao
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, PR China.,College of Veterinary Medicine, Jilin University, Changchun, China
| | - X Xu
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, PR China.,College of Veterinary Medicine, Jilin University, Changchun, China
| | - P Zhang
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - J Wang
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, PR China.,College of Veterinary Medicine, Jilin University, Changchun, China
| | - G Li
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Q Lv
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, PR China.,College of Veterinary Medicine, Jilin University, Changchun, China
| | - X Niu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - H Liu
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, PR China
| |
Collapse
|
18
|
Wang G, Gao Y, Wu X, Gao X, Zhang M, Liu H, Fang T. Inhibitory Effect of Piceatannol on Streptococcus suis Infection Both in vitro and in vivo. Front Microbiol 2020; 11:593588. [PMID: 33329477 PMCID: PMC7728846 DOI: 10.3389/fmicb.2020.593588] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/04/2020] [Indexed: 11/18/2022] Open
Abstract
Suilysin (SLY) plays a critical role in Streptococcus suis infections making it an ideal target to the combat infection caused by this pathogen. In the present study, we found that piceatannol (PN), a natural compound, inhibits pore-formation by blocking the oligomerization of SLY without affecting the growth of S. suis and the expression of SLY. Furthermore, PN alleviated the J774 cell damage and the expression of the inflammatory cytokine tumor necrosis factor-α (TNF-α) and interleukin-1α (IL-1β) induced by S. suis in vitro. The computational biology and biochemistry results indicated that PN binds to the joint region of D2 and D4 in SLY, and Asn57, Pro58, Pro59, Glu76, Ile379, Glu380, and Glu418 were critical residues involved in the binding. The binding effect between PN and SLY hindered the SLY monomers from forming the oligomers, thereby weakening the hemolytic activity of SLY. This mechanism was also verified by hemolysis analysis and analysis of KA formation after site-specific mutagenesis. Furthermore, PN protected mice from S. suis infections by reducing bacterial colony formation and the inflammatory response in target organs in vivo. These results indicate that PN is a feasible drug candidate to combat S. suis infections.
Collapse
Affiliation(s)
- Guizhen Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China.,College of Food Engineering, Jilin Engineering Normal University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yawen Gao
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiuhua Wu
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Xiue Gao
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Min Zhang
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Hongmei Liu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Tianqi Fang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
19
|
Zeng Z, Xie J, Luo G, Tao Z, Zhang Q. Host-guest interaction of cucurbit[8]uril with oroxin A and its effect on the properties of oroxin A. Beilstein J Org Chem 2020; 16:2332-2337. [PMID: 33029251 PMCID: PMC7522457 DOI: 10.3762/bjoc.16.194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/14/2020] [Indexed: 01/06/2023] Open
Abstract
In this study, we investigated the host-guest interactions between oroxin A (OA) and cucurbit[8]uril (Q[8]) using 1H NMR, MS, UV-vis and IR spectroscopy. The results showed that OA and Q[8] formed an inclusion compound (OA@Q[8]) with a molar ratio of 1:1 and a binding constant of 1.299 × 107 L·mol-1. In addition, the effect of Q[8] on the properties of OA was investigated through comparative experiments. The solubility of OA in water increased 22.47-fold when the concentration of Q[8] was 1 × 10-4 mol·L-1. Q[8] hardly affected the antioxidant capacity of OA, while the cumulative release of OA in gastric juice increased 2.3-fold after forming the inclusion compound with Q[8].
Collapse
Affiliation(s)
- Zhishu Zeng
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, No. 2708, South Section of Huaxi Avenue, Huaxi, Guiyang 550025, China
| | - Jun Xie
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, No. 2708, South Section of Huaxi Avenue, Huaxi, Guiyang 550025, China
| | - Guangyan Luo
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, No. 2708, South Section of Huaxi Avenue, Huaxi, Guiyang 550025, China
| | - Zhu Tao
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, No. 2708, South Section of Huaxi Avenue, Huaxi, Guiyang 550025, China
| | - Qianjun Zhang
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, No. 2708, South Section of Huaxi Avenue, Huaxi, Guiyang 550025, China
| |
Collapse
|
20
|
Inhibition of interaction between Staphylococcus aureus α-hemolysin and erythrocytes membrane by hydrolysable tannins: structure-related activity study. Sci Rep 2020; 10:11168. [PMID: 32636484 PMCID: PMC7341856 DOI: 10.1038/s41598-020-68030-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/11/2020] [Indexed: 01/17/2023] Open
Abstract
The objective of the study was a comparative analysis of the antihemolytic activity against two Staphylococcus aureus strains (8325-4 and NCTC 5655) as well as α-hemolysin and of the membrane modifying action of four hydrolysable tannins with different molecular mass and flexibility: 3,6-bis-O-di-O-galloyl-1,2,4-tri-O-galloyl-β-d-glucose (T1), 1,2,3,4,5-penta-O-galloyl-β-d-glucose (T2), 3-O-galloyl-1,2-valoneoyl-β-d-glucose (T3) and 1,2-di-O-galloyl-4,6-valoneoyl-β-d-glucose (T4). We showed that all the compounds studied manifested antihemolytic effects in the range of 5–50 µM concentrations. However, the degree of the reduction of hemolysis by the investigated tannins was not uniform. A valoneoyl group—containing compounds (T3 and T4) were less active. Inhibition of the hemolysis induced by α-hemolysin was also noticed on preincubated with the tannins and subsequently washed erythrocytes. In this case the efficiency again depended on the tannin structure and could be represented by the following order: T1 > T2 > T4 > T3. We also found a relationship between the degree of antihemolytic activity of the tannins studied and their capacity to increase the ordering parameter of the erythrocyte membrane outer layer and to change zeta potential. Overall, our study showed a potential of the T1 and T2 tannins as anti-virulence agents. The results of this study using tannins with different combinations of molecular mass and flexibility shed additional light on the role of tannin structure in activity manifestation.
Collapse
|
21
|
Marchetti M, De Bei O, Bettati S, Campanini B, Kovachka S, Gianquinto E, Spyrakis F, Ronda L. Iron Metabolism at the Interface between Host and Pathogen: From Nutritional Immunity to Antibacterial Development. Int J Mol Sci 2020; 21:E2145. [PMID: 32245010 PMCID: PMC7139808 DOI: 10.3390/ijms21062145] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 02/08/2023] Open
Abstract
Nutritional immunity is a form of innate immunity widespread in both vertebrates and invertebrates. The term refers to a rich repertoire of mechanisms set up by the host to inhibit bacterial proliferation by sequestering trace minerals (mainly iron, but also zinc and manganese). This strategy, selected by evolution, represents an effective front-line defense against pathogens and has thus inspired the exploitation of iron restriction in the development of innovative antimicrobials or enhancers of antimicrobial therapy. This review focuses on the mechanisms of nutritional immunity, the strategies adopted by opportunistic human pathogen Staphylococcus aureus to circumvent it, and the impact of deletion mutants on the fitness, infectivity, and persistence inside the host. This information finally converges in an overview of the current development of inhibitors targeting the different stages of iron uptake, an as-yet unexploited target in the field of antistaphylococcal drug discovery.
Collapse
Affiliation(s)
- Marialaura Marchetti
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
| | - Omar De Bei
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Stefano Bettati
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
- National Institute of Biostructures and Biosystems, 00136 Rome, Italy
| | - Barbara Campanini
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Sandra Kovachka
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Luca Ronda
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
| |
Collapse
|
22
|
Fox D, Mathur A, Xue Y, Liu Y, Tan WH, Feng S, Pandey A, Ngo C, Hayward JA, Atmosukarto II, Price JD, Johnson MD, Jessberger N, Robertson AAB, Burgio G, Tscharke DC, Fox EM, Leyton DL, Kaakoush NO, Märtlbauer E, Leppla SH, Man SM. Bacillus cereus non-haemolytic enterotoxin activates the NLRP3 inflammasome. Nat Commun 2020; 11:760. [PMID: 32029733 PMCID: PMC7005308 DOI: 10.1038/s41467-020-14534-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
Inflammasomes are important for host defence against pathogens and homeostasis with commensal microbes. Here, we show non-haemolytic enterotoxin (NHE) from the neglected human foodborne pathogen Bacillus cereus is an activator of the NLRP3 inflammasome and pyroptosis. NHE is a non-redundant toxin to haemolysin BL (HBL) despite having a similar mechanism of action. Via a putative transmembrane region, subunit C of NHE initiates binding to the plasma membrane, leading to the recruitment of subunit B and subunit A, thus forming a tripartite lytic pore that is permissive to efflux of potassium. NHE mediates killing of cells from multiple lineages and hosts, highlighting a versatile functional repertoire in different host species. These data indicate that NHE and HBL operate synergistically to induce inflammation and show that multiple virulence factors from the same pathogen with conserved function and mechanism of action can be exploited for sensing by a single inflammasome.
Collapse
Affiliation(s)
- Daniel Fox
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Anukriti Mathur
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Yansong Xue
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Yunqi Liu
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Wei Hong Tan
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Shouya Feng
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Abhimanu Pandey
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Chinh Ngo
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Jenni A Hayward
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Ines I Atmosukarto
- Lipotek Pty Ltd. The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Jason D Price
- Lipotek Pty Ltd. The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Matthew D Johnson
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Nadja Jessberger
- Department of Veterinary Sciences, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | - Avril A B Robertson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Gaetan Burgio
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - David C Tscharke
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Edward M Fox
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Denisse L Leyton
- Research School of Biology, The Australian National University, Canberra, Australia.,Medical School, The Australian National University, Canberra, Australia
| | - Nadeem O Kaakoush
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Erwin Märtlbauer
- Department of Veterinary Sciences, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | - Stephen H Leppla
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Si Ming Man
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
| |
Collapse
|
23
|
Oroxin B Induces Apoptosis by Down-Regulating MicroRNA-221 Resulting in the Inactivation of the PTEN/PI3K/AKT Pathway in Liver Cancer. Molecules 2019; 24:molecules24234384. [PMID: 31801250 PMCID: PMC6930563 DOI: 10.3390/molecules24234384] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
This study aims to investigate the anticancer effect of Oroxin B (OB) both in vitro and in vivo, and the molecular mechanism involved in microRNA-221 and the PI3K/Akt/PTEN pathway through modulation of apoptosis in Hepatocellular carcinoma (HCC). DEN-induced rats and HepG2 cells based on the microfluidic chip were employed, while the mRNA and protein expression of microRNA-221, PI3K, p-Akt and PTEN were evaluated by RT-PCR and Western blot analysis. Based on Microfluidic Chip and DEN-induced rat model, OB effectively exerts anti-liver cancer effect both in vitro and in vivo, and the expression of miR-221 in OB treated groups was significantly lower than that in the control group (** p < 0.01). The RT-PCR and Western blot results suggested the PI3K mRNA and protein in OB treated groups were both lower than those in control group and indicated the overexpression of PTEN. Therefore, OB effectively exerts anticancer effects by positively regulating the PTEN gene and then inactivating the PI3K/Akt signaling pathway through down-regulating the expression of the microRNA-221, thereby inducing apoptosis of liver cancer cells. This study offers a theoretical evidence for further development and clinical guidance of OB as an anti-tumor agent.
Collapse
|
24
|
Omersa N, Podobnik M, Anderluh G. Inhibition of Pore-Forming Proteins. Toxins (Basel) 2019; 11:E545. [PMID: 31546810 PMCID: PMC6784129 DOI: 10.3390/toxins11090545] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/27/2019] [Accepted: 09/10/2019] [Indexed: 12/16/2022] Open
Abstract
Perforation of cellular membranes by pore-forming proteins can affect cell physiology, tissue integrity, or immune response. Since many pore-forming proteins are toxins or highly potent virulence factors, they represent an attractive target for the development of molecules that neutralize their actions with high efficacy. There has been an assortment of inhibitors developed to specifically obstruct the activity of pore-forming proteins, in addition to vaccination and antibiotics that serve as a plausible treatment for the majority of diseases caused by bacterial infections. Here we review a wide range of potential inhibitors that can specifically and effectively block the activity of pore-forming proteins, from small molecules to more specific macromolecular systems, such as synthetic nanoparticles, antibodies, antibody mimetics, polyvalent inhibitors, and dominant negative mutants. We discuss their mechanism of inhibition, as well as advantages and disadvantages.
Collapse
Affiliation(s)
- Neža Omersa
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| |
Collapse
|
25
|
Staphylococcus aureus Toxins: From Their Pathogenic Roles to Anti-virulence Therapy Using Natural Products. BIOTECHNOL BIOPROC E 2019. [DOI: 10.1007/s12257-019-0059-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
26
|
Lin SJ, Chen YF, Hsu KC, Chen YL, Ko TP, Lo CF, Wang HC, Wang HC. Structural Insights to the Heterotetrameric Interaction between the Vibrio parahaemolyticus PirA vp and PirB vp Toxins and Activation of the Cry-Like Pore-Forming Domain. Toxins (Basel) 2019; 11:toxins11040233. [PMID: 31013623 PMCID: PMC6520838 DOI: 10.3390/toxins11040233] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/16/2019] [Accepted: 04/19/2019] [Indexed: 02/06/2023] Open
Abstract
Acute hepatopancreatic necrosis disease (AHPND) is a newly emergent penaeid shrimp disease which can cause 70-100% mortality in Penaeus vannamei and Penaeus monodon, and has resulted in enormous economic losses since its appearance. AHPND is caused by the specific strains of Vibrio parahaemolyticus that harbor the pVA1 plasmid and express PirAvp and PirBvp toxins. These two toxins have been reported to form a binary complex. When both are present, they lead to the death of shrimp epithelial cells in the hepatopancreas and cause the typical histological symptoms of AHPND. However, the binding mode of PirAvp and PirBvp has not yet been determined. Here, we used isothermal titration calorimetry (ITC) to measure the binding affinity of PirAvp and PirBvp. Since the dissociation constant (Kd = 7.33 ± 1.20 μM) was considered too low to form a sufficiently stable complex for X-ray crystallographic analysis, we used alternative methods to investigate PirAvp-PirBvp interaction, first by using gel filtration to evaluate the molecular weight of the PirAvp/PirBvp complex, and then by using cross-linking and hydrogen-deuterium exchange (HDX) mass spectrometry to further understand the interaction interface between PirAvp and PirBvp. Based on these results, we propose a heterotetrameric interaction model of this binary toxin complex. This model provides insight of how conformational changes might activate the PirBvp N-terminal pore-forming domain and should be helpful for devising effective anti-AHPND strategies in the future.
Collapse
Affiliation(s)
- Shin-Jen Lin
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan.
| | - Yi-Fan Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan.
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
- Biomedical Commercialization Center, Taipei Medical University, Taipei 110, Taiwan.
| | - Yun-Ling Chen
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan.
| | - Tzu-Ping Ko
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan.
| | - Chu-Fang Lo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan.
- International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan 701, Taiwan.
| | - Han-Ching Wang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan.
- International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan 701, Taiwan.
| | - Hao-Ching Wang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan.
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
- International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
27
|
Zhao Y, Xu Z, Wang T, Li Y, Yang L, Liu S, Shi R, Ma Y. Simultaneous quantitation of 23 bioactive compounds in Tanreqing capsule by high‐performance liquid chromatography electrospray ionization tandem mass spectrometry. Biomed Chromatogr 2019; 33:e4531. [DOI: 10.1002/bmc.4531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 02/23/2019] [Accepted: 03/01/2019] [Indexed: 01/17/2023]
Affiliation(s)
- Yining Zhao
- Department of PharmacologyShanghai University of Traditional Chinese Medicine Shanghai China
| | - Zhangyao Xu
- Department of PharmacologyShanghai University of Traditional Chinese Medicine Shanghai China
| | - Tianming Wang
- Department of PharmacologyShanghai University of Traditional Chinese Medicine Shanghai China
| | - Yuanyuan Li
- Department of PharmacologyShanghai University of Traditional Chinese Medicine Shanghai China
| | - Li Yang
- Centre for Traditional Chinese Medicine of Complexity SystemsShanghai University of Traditional Chinese Medicine Shanghai China
| | - Shaoyong Liu
- Shanghai Kai Bao Pharmaceutical CO. Ltd Shanghai China
| | - Rong Shi
- Department of PharmacologyShanghai University of Traditional Chinese Medicine Shanghai China
| | - Yueming Ma
- Department of PharmacologyShanghai University of Traditional Chinese Medicine Shanghai China
| |
Collapse
|
28
|
Divyakolu S, Chikkala R, Ratnakar KS, Sritharan V. Hemolysins of <i>Staphylococcus aureus</i>—An Update on Their Biology, Role in Pathogenesis and as Targets for Anti-Virulence Therapy. ACTA ACUST UNITED AC 2019. [DOI: 10.4236/aid.2019.92007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
29
|
Niu X, Yu Y, Guo H, Yang Y, Wang G, Sun L, Gao Y, Yu Z, Wang H. Molecular modeling reveals the inhibition mechanism and binding mode of ursolic acid to TLR4-MD2. COMPUT THEOR CHEM 2018. [DOI: 10.1016/j.comptc.2017.11.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
30
|
Insight into the novel inhibition mechanism of apigenin to Pneumolysin by molecular modeling. Chem Phys Lett 2017. [DOI: 10.1016/j.cplett.2017.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Investigation of the inhibition effect and mechanism of myricetin to Suilysin by molecular modeling. Sci Rep 2017; 7:11748. [PMID: 28924148 PMCID: PMC5603505 DOI: 10.1038/s41598-017-12168-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/04/2017] [Indexed: 01/27/2023] Open
Abstract
In the present study, the inhibitory effect and mechanism of myricetin, a natural flavonoid compound, in relation to Suilysin (SLY) were investigated through molecular dynamics simulations, mutational analysis and fluorescence-quenching assays. Myricetin is a potential inhibitor that does not exhibit antimicrobial activity but has been shown to inhibit SLY cytotoxicity. Molecular dynamics simulations and mutational analysis revealed that myricetin binds directly to SLY in the gap between domains 2 and 3, an important region for oligomerization and pore formation. The results of principal component analysis (PCA) indicated that the binding of myricetin in this gap region restricts the conformational transition of SLY from a monomer to an oligomer, thereby counteracting the haemolytic activity of SLY. This mechanism was verified using a haemolysis assay. These results demonstrated that myricetin is a strong candidate as a novel therapeutic agent for the treatment of Streptococcus suis infections.
Collapse
|
32
|
Li H, Zhao X, Deng X, Wang J, Song M, Niu X, Peng L. Insights into structure and activity of natural compound inhibitors of pneumolysin. Sci Rep 2017; 7:42015. [PMID: 28165051 PMCID: PMC5292752 DOI: 10.1038/srep42015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 01/05/2017] [Indexed: 01/03/2023] Open
Abstract
Pneumolysin is the one of the major virulence factor of the bacterium Streptococcus pneumoniae. In previous report, it is shown that β-sitosterol, a natural compound without antimicrobial activity, is a potent antagonist of pneumolysin. Here, two new pneumolysin natural compound inhibitors, with differential activity, were discovered via haemolysis assay. To explore the key factor of the conformation for the inhibition activity, the interactions between five natural compound inhibitors with differential activity and pneumolysin were reported using molecular modelling, the potential of mean force profiles. Interestingly, it is found that incorporation of the single bond (C22-C23-C24-C25) to replace the double bond (hydrocarbon sidechain) improved the anti-haemolytic activity. In view of the molecular modelling, binding of the five inhibitors to the conserved loop region (Val372, Leu460, and Tyr461) of the cholesterol binding sites led to stable complex systems, which was consistent with the result of β-sitosterol. Owing to the single bond (C22-C23-C24-C25), campesterol and brassicasterol could form strong interactions with Val372 and show higher anti-haemolytic activity, which indicated that the single bond (C22-C23-C24-C25) in inhibitors was required for the anti-haemolytic activity. Overall, the current molecular modelling work provides a starting point for the development of rational design and higher activity pneumolysin inhibitors.
Collapse
Affiliation(s)
- Hongen Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaoran Zhao
- Department of Respiratory Medicine, The First Hospital of Jilin University, Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuming Deng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianfeng Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Meng Song
- Department of Respiratory Medicine, The First Hospital of Jilin University, Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaodi Niu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Liping Peng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
33
|
Saeloh D, Wenzel M, Rungrotmongkol T, Hamoen LW, Tipmanee V, Voravuthikunchai SP. Effects of rhodomyrtone on Gram-positive bacterial tubulin homologue FtsZ. PeerJ 2017; 5:e2962. [PMID: 28168121 PMCID: PMC5292029 DOI: 10.7717/peerj.2962] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/05/2017] [Indexed: 12/15/2022] Open
Abstract
Rhodomyrtone, a natural antimicrobial compound, displays potent activity against many Gram-positive pathogenic bacteria, comparable to last-defence antibiotics including vancomycin and daptomycin. Our previous studies pointed towards effects of rhodomyrtone on the bacterial membrane and cell wall. In addition, a recent molecular docking study suggested that the compound could competitively bind to the main bacterial cell division protein FtsZ. In this study, we applied a computational approach (in silico), in vitro, and in vivo experiments to investigate molecular interactions of rhodomyrtone with FtsZ. Using molecular simulation, FtsZ conformational changes were observed in both (S)- and (R)-rhodomyrtone binding states, compared with the three natural states of FtsZ (ligand-free, GDP-, and GTP-binding states). Calculations of free binding energy showed a higher affinity of FtsZ to (S)-rhodomyrtone (−35.92 ± 0.36 kcal mol−1) than the GDP substrate (−23.47 ± 0.25 kcal mol−1) while less affinity was observed in the case of (R)-rhodomyrtone (−18.11 ± 0.11 kcal mol−1). In vitro experiments further revealed that rhodomyrtone reduced FtsZ polymerization by 36% and inhibited GTPase activity by up to 45%. However, the compound had no effect on FtsZ localization in Bacillus subtilis at inhibitory concentrations and cells also did not elongate after treatment. Higher concentrations of rhodomyrtone did affect localization of FtsZ and also affected localization of its membrane anchor proteins FtsA and SepF, showing that the compound did not specifically inhibit FtsZ but rather impaired multiple divisome proteins. Furthermore, a number of cells adopted a bean-like shape suggesting that rhodomyrtone possibly possesses further targets involved in cell envelope synthesis and/or maintenance.
Collapse
Affiliation(s)
- Dennapa Saeloh
- Excellence Research Laboratory on Natural Products, Faculty of Science and Natural Product Research Center of Excellence, Prince of Songkla University, Hat Yai, Thailand; Department of Microbiology, Faculty of Science, Prince of Songkla University, Hat Yai, Thailand
| | - Michaela Wenzel
- Bacterial Cell Biology, Swammerdam Institute for Life Sciences, University of Amsterdam , Amsterdam , Netherlands
| | - Thanyada Rungrotmongkol
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand; Center of Innovative Nanotechnology, Chulalongkorn University, Bongkok, Thailand
| | - Leendert Willem Hamoen
- Bacterial Cell Biology, Swammerdam Institute for Life Sciences, University of Amsterdam , Amsterdam , Netherlands
| | - Varomyalin Tipmanee
- Excellence Research Laboratory on Natural Products, Faculty of Science and Natural Product Research Center of Excellence, Prince of Songkla University, Hat Yai, Thailand; Department of Biomedical Science, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Supayang Piyawan Voravuthikunchai
- Excellence Research Laboratory on Natural Products, Faculty of Science and Natural Product Research Center of Excellence, Prince of Songkla University, Hat Yai, Thailand; Department of Microbiology, Faculty of Science, Prince of Songkla University, Hat Yai, Thailand
| |
Collapse
|
34
|
Silva LN, Zimmer KR, Macedo AJ, Trentin DS. Plant Natural Products Targeting Bacterial Virulence Factors. Chem Rev 2016; 116:9162-236. [PMID: 27437994 DOI: 10.1021/acs.chemrev.6b00184] [Citation(s) in RCA: 286] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Decreased antimicrobial efficiency has become a global public health issue. The paucity of new antibacterial drugs is evident, and the arsenal against infectious diseases needs to be improved urgently. The selection of plants as a source of prototype compounds is appropriate, since plant species naturally produce a wide range of secondary metabolites that act as a chemical line of defense against microorganisms in the environment. Although traditional approaches to combat microbial infections remain effective, targeting microbial virulence rather than survival seems to be an exciting strategy, since the modulation of virulence factors might lead to a milder evolutionary pressure for the development of resistance. Additionally, anti-infective chemotherapies may be successfully achieved by combining antivirulence and conventional antimicrobials, extending the lifespan of these drugs. This review presents an updated discussion of natural compounds isolated from plants with chemically characterized structures and activity against the major bacterial virulence factors: quorum sensing, bacterial biofilms, bacterial motility, bacterial toxins, bacterial pigments, bacterial enzymes, and bacterial surfactants. Moreover, a critical analysis of the most promising virulence factors is presented, highlighting their potential as targets to attenuate bacterial virulence. The ongoing progress in the field of antivirulence therapy may therefore help to translate this promising concept into real intervention strategies in clinical areas.
Collapse
Affiliation(s)
- Laura Nunes Silva
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil
| | - Karine Rigon Zimmer
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre , Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Alexandre José Macedo
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil.,Instituto Nacional do Semiárido , Campina Grande, Paraı́ba 58429-970, Brazil
| | - Danielle Silva Trentin
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil
| |
Collapse
|
35
|
Kong C, Neoh HM, Nathan S. Targeting Staphylococcus aureus Toxins: A Potential form of Anti-Virulence Therapy. Toxins (Basel) 2016; 8:toxins8030072. [PMID: 26999200 PMCID: PMC4810217 DOI: 10.3390/toxins8030072] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/03/2016] [Accepted: 03/10/2016] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen and the leading cause of a wide range of severe clinical infections. The range of diseases reflects the diversity of virulence factors produced by this pathogen. To establish an infection in the host, S. aureus expresses an inclusive set of virulence factors such as toxins, enzymes, adhesins, and other surface proteins that allow the pathogen to survive under extreme conditions and are essential for the bacteria’s ability to spread through tissues. Expression and secretion of this array of toxins and enzymes are tightly controlled by a number of regulatory systems. S. aureus is also notorious for its ability to resist the arsenal of currently available antibiotics and dissemination of various multidrug-resistant S. aureus clones limits therapeutic options for a S. aureus infection. Recently, the development of anti-virulence therapeutics that neutralize S. aureus toxins or block the pathways that regulate toxin production has shown potential in thwarting the bacteria’s acquisition of antibiotic resistance. In this review, we provide insights into the regulation of S. aureus toxin production and potential anti-virulence strategies that target S. aureus toxins.
Collapse
Affiliation(s)
- Cin Kong
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor Darul Ehsan, Malaysia.
| | - Hui-min Neoh
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur, Malaysia.
| | - Sheila Nathan
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
36
|
Zhao X, Li H, Wang J, Guo Y, Liu B, Deng X, Niu X. Verbascoside Alleviates Pneumococcal Pneumonia by Reducing Pneumolysin Oligomers. Mol Pharmacol 2016; 89:376-87. [PMID: 26700563 DOI: 10.1124/mol.115.100610] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/18/2015] [Indexed: 02/14/2025] Open
Abstract
Pneumolysin (PLY), an essential virulence factor of Streptococcus pneumoniae (pneumococcus), can penetrate the physical defenses of the host and possesses inflammatory properties. The vital role PLY plays in pneumococcus pathogenesis makes this virulence factor one of the most promising targets for the treatment of pneumococcal infection. Verbascoside (VBS) is an agent that does not exhibit bacteriostatic activity but has been shown to inhibit PLY-mediated cytotoxicity. The results from molecular dynamics simulations and mutational analysis indicated that VBS binds to the cleft between domains 3 and 4 of PLY, thereby blocking PLY's oligomerization and counteracting its hemolytic activity. Moreover, VBS can effectively alleviate PLY-mediated human alveolar epithelial (A549) cell injury, and treatment with VBS provides significant protection against lung damage and reduces mortality in a pneumococcal pneumonia murine model. Our results demonstrate that VBS is a strong candidate as a novel therapeutic in the treatment of Streptococcus pneumoniae infection.
Collapse
Affiliation(s)
- Xiaoran Zhao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.)
| | - Hongen Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.)
| | - Jianfeng Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.)
| | - Yan Guo
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.)
| | - Bowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.)
| | - Xuming Deng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.).
| | - Xiaodi Niu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.).
| |
Collapse
|
37
|
He J, Du L, Bao M, Zhang B, Qian H, Zhou Q, Cao Z. Oroxin A inhibits breast cancer cell growth by inducing robust endoplasmic reticulum stress and senescence. Anticancer Drugs 2016; 27:204-215. [PMID: 26599214 DOI: 10.1097/cad.0000000000000318] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Breast cancer is a major cause of cancer death among women. Although various anticancer drugs have been used in clinics, drugs that are effective against advanced and metastatic breast cancer are still lacking and in great demand. In this study, we found that oroxin A, an active component isolated from the herb Oroxylum indicum (L.) Kurz, effectively inhibited the growth of human breast cancer cells MDA-MB-231 and MCF7 by inducing endoplasmic reticulum (ER) stress-mediated senescence. Oroxin A caused breast cancer cell cycle arrest at the G2/M stage, and reorganization of microtubules and actin cytoskeleton accompanied by a decrease in cellular mitosis. ER-specific probe ER-Tracker Red and confocal microscope imaging showed that ER-Tracker Red-positive cells increased in an oroxin A dosage-dependent manner. In addition, oroxin A increased cell population with high β-Gal activity and SAHF-positive staining; these data suggest that oroxin A induces breast cancer cell ER stress and senescence. Mechanistic studies showed that oroxin A led to a significant increase in intracellular reactive oxygen species levels, promoted expression of ER stress markers ATF4 and GRP78, and increased the phosphorylation of a key stress-response signaling protein p38, resulting in an ER stress-mediated senescence. Taken together, our data indicate that oroxin A exerts its antibreast cancer effects by inducing ER stress-mediated senescence, activating the key stress p38 signaling pathway, and increasing key ER stress genes ATF4 and GRP78 expression levels.
Collapse
Affiliation(s)
- Jun He
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Ministry of Health, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
38
|
Peraro MD, van der Goot FG. Pore-forming toxins: ancient, but never really out of fashion. Nat Rev Microbiol 2015; 14:77-92. [DOI: 10.1038/nrmicro.2015.3] [Citation(s) in RCA: 476] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
39
|
Melo MCA, Teixeira LR, Pol-Fachin L, Rodrigues CG. Inhibition of the hemolytic activity caused by Staphylococcus aureus alpha-hemolysin through isatin-Schiff copper(II) complexes. FEMS Microbiol Lett 2015; 363:fnv207. [PMID: 26519261 DOI: 10.1093/femsle/fnv207] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2015] [Indexed: 02/01/2023] Open
Abstract
A great number of pathogens secrete pore-forming proteins during infection. Such molecules, from either bacterial or viral origin, are considered important virulence factors, which makes them attractive targets in the study of new therapeutic agents. Thus, the inhibitory activity of isatin-Schiff base copper(II) complexes was evaluated against membrane damage activity of Staphylococcus aureus α-hemolysin (α-HL). For this purpose, a standard hemolysis assay with rabbit erythrocytes and micromolar concentrations of the compounds was employed. Additionally, planar artificial lipid membranes with a single α-HL ion channel and molecular docking studies were used to elucidate the molecular mechanism of the complexes. Accordingly, the compounds were observed to possess a significant anti-hemolytic activity, capable of interacting with the constriction region of α-HL channel and blocking it in a potential dependent manner. Based on these results, it is expected that such isatin-Schiff base Copper(II) complexes may be employed as cotherapeutic agents for the treatment of staphylococcal infections.
Collapse
Affiliation(s)
- Maria C A Melo
- Laboratory of Membrane Biophysics and Stem Cell Dr. Oleg Krasilnikov, Department of Biophysics and Radiobiology, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Luciana R Teixeira
- Laboratory of Membrane Biophysics and Stem Cell Dr. Oleg Krasilnikov, Department of Biophysics and Radiobiology, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Laercio Pol-Fachin
- Biomaterials Laboratory, Department of Fundamental Chemistry, Federal University of Pernambuco, Recife, Pernambuco, Brazil Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Claudio G Rodrigues
- Laboratory of Membrane Biophysics and Stem Cell Dr. Oleg Krasilnikov, Department of Biophysics and Radiobiology, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| |
Collapse
|
40
|
Wang J, Zhou X, Liu S, Li G, Shi L, Dong J, Li W, Deng X, Niu X. Morin hydrate attenuates Staphylococcus aureus virulence by inhibiting the self-assembly of α-hemolysin. J Appl Microbiol 2015; 118:753-63. [PMID: 25564958 DOI: 10.1111/jam.12743] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/17/2014] [Accepted: 12/23/2014] [Indexed: 11/26/2022]
Abstract
AIMS To investigate the mechanism by which morin hydrate inhibits the haemolytic activity of α-hemolysin (Hla), a channel-forming toxin that is important for the pathogenesis of disease in experimental animals, and its therapeutic effect against Staphylococcus aureus pneumonia in a mouse model. METHODS AND RESULTS The results from the in vitro (haemolysis, western blot and cytotoxicity assays) and in vivo (mouse model of intranasal lung infection) experiments indicated that morin hydrate, a natural compound with little anti-Staph. aureus activity, could effectively antagonize the cytolytic activity of Hla, alleviate human lung cell injury, and protect against mortality of Staph. aureus pneumonia in a mouse model of infection. Molecular dynamics simulations, free energy calculations and mutagenesis assays were further employed to determine the catalytic mechanism of inhibition, which indicated that a direct binding of morin to the 'Stem' domain of Hla (residues I107 and T109) and the concomitant change in conformation led to the inhibition of the self-assembly of the heptameric transmembrane pore, thus inhibiting the biological activity of Hla for cell lysis. CONCLUSIONS Morin inhibited Staph. aureus virulence via inhibiting the haemolytic activity of α-hemolysin. SIGNIFICANCE AND IMPACT OF THE STUDY These findings suggested that morin is a promising candidate for the development of anti-virulence therapeutic agents for the treatment of Staph. aureus infections.
Collapse
Affiliation(s)
- J Wang
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chen H, He G, Li C, Dong L, Xie X, Wu J, Gao Y, Zhou J. Development of a Concise Synthetic Approach to Access Oroxin A. RSC Adv 2014; 4:45151-45154. [PMID: 25431654 DOI: 10.1039/c4ra08573f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A novel environment-friendly method to access bioactive oroxin A through a one-pot/two-step process from naturally abundant and inexpensive baicalin is described. The procedure presented here has several advantages including clean, one-pot, synthetic ease, and large-scale feasibility. This work also provides a model strategy for rapid and diverse access to natural molecules sharing the common skeleton of this family.
Collapse
Affiliation(s)
- Haijun Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108,China,; Tel: +86 591 22866234; (H. Chen). ; Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States, Fax: +1 (409) 772-9648; Tel: +1 (409) 772-9748; (J. Zhou)
| | - Guihua He
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108,China,; Tel: +86 591 22866234; (H. Chen)
| | - Cailong Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108,China,; Tel: +86 591 22866234; (H. Chen)
| | - Longrong Dong
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108,China,; Tel: +86 591 22866234; (H. Chen)
| | - Xiaobo Xie
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108,China,; Tel: +86 591 22866234; (H. Chen)
| | - Jianlei Wu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108,China,; Tel: +86 591 22866234; (H. Chen)
| | - Yu Gao
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108,China,; Tel: +86 591 22866234; (H. Chen)
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States, ; Tel: +1 (409) 772-9748; (J. Zhou)
| |
Collapse
|