1
|
Wang SH, Kuo BJ, Ho TC, Wan SW, Yen KL, Huang PH, Perng OGC, Chen PL, Chien YW, Lo YC. Lambda-free light chain: A serum marker of dengue disease via NS3 protease-mediated antibody cleavage. Virulence 2023; 14:2279355. [PMID: 37927064 PMCID: PMC10766417 DOI: 10.1080/21505594.2023.2279355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023] Open
Abstract
Dengue poses a significant global public health threat, with diverse clinical manifestations due to complex interactions between the host and the pathogen. Recent reports have highlighted elevated serum-free light chain (FLC) levels in viral infectious diseases. Hence, our study aimed to investigate serum FLC levels in dengue patients. The findings revealed elevated serum λ FLCs, which were associated with the severity of dengue. Receiver operating characteristic curve (ROC) analysis demonstrated that λ FLCs may serve as a serum marker for identifying dengue disease (AUC: 0.7825, sensitivity: 80, specificity: 71.43) and classifying severe dengue (AUC: 0.8102, sensitivity: 75, specificity: 79.52). The viral protease, Dengue virus (DENV) nonstructural protein 3 (NS3), acts as a protease that cleaves viral polyproteins as well as host substrates. Therefore, we proposed that antibodies might be potential targets of NS3 protease, leading to an increase in FLCs. LC/MS-MS analysis confirmed that λ FLCs were the predominant products after antibody degradation by NS3 protease. Additionally, purified NS3 protease cleaved both human IgG and DENV2-neutralizing antibodies, resulting in the presence of λ FLCs. Moreover, NS3 protease administration in vitro led to a reduction in the neutralizing efficacy of DENV2-neutralizing antibodies. In summary, the elevated serum λ FLC levels effectively differentiate dengue patients from healthy individuals and identify severe dengue. Furthermore, the elevation of serum λ FLCs is, at least in part, mediated through NS3 protease-mediated antibody cleavage. These findings provide new insights for developing diagnostic tools and understanding the pathogenesis of DENV infection.
Collapse
Affiliation(s)
- Sheng-Hsuan Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bai-Jiun Kuo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Chuan Ho
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Wen Wan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Ko-Lun Yen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Hui Huang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Oscar Guey Chuen Perng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Po-Lin Chen
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- Center for Infection Control, National Cheng Kung University Medical College and Hospital, Tainan, Taiwan
| | - Yu-Wen Chien
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Occupational and Environmental Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yu-Chih Lo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
2
|
Chen YY, Li BP, Wang JF, Wang Y, Luo SS, Lin RJ, Liao XW, Chen JQ. Investigating the prognostic and predictive value of the type II cystatin genes in gastric cancer. BMC Cancer 2023; 23:1122. [PMID: 37978366 PMCID: PMC10657128 DOI: 10.1186/s12885-023-11550-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 10/19/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Accumulating evidence indicates that type II cystatin (CST) genes play a pivotal role in several tumor pathological processes, thereby affecting all stages of tumorigenesis and tumor development. However, the prognostic and predictive value of type II CST genes in GC has not yet been investigated. METHODS The present study evaluated the expression and prognostic value of type II CST genes in GC by using The Cancer Genome Atlas (TCGA) database and the Kaplan-Meier plotter (KM plotter) online database. The type II CST genes related to the prognosis of GC were then screened out. We then validated the expression and prognostic value of these genes by immunohistochemistry. We also used Database for Annotation, Visualization, and Integrated Discovery (DAVID), Gene Multiple Association Network Integration Algorithm (GeneMANIA), Search Tool for the Retrieval of Interacting Genes/Proteins (STRING), nomogram, genome-wide co-expression analysis, and other bioinformatics tools to analyze the value of type II CST genes in GC and the underlying mechanism. RESULTS The data from the TCGA database and the KM plotter online database showed that high expression of CST2 and CST4 was associated with the overall survival (OS) of patients with GC. The immunohistochemical expression analysis showed that patients with high expression of CST4 in GC tissues have a shorter OS than those with low expression of CST4 (HR = 1.85,95%CI: 1.13-3.03, P = 0.015). Multivariate Cox regression analysis confirmed that the high expression level of CST4 was an independent prognostic risk factor for OS. CONCLUSIONS Our findings suggest that CST4 could serve as a tumor marker that affects the prognosis of GC and could be considered as a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Ye-Yang Chen
- Department of General Surgery, The First People's Hospital of Yulin, Yulin, China
| | - Bo-Pei Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun-Fu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ye Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shan-Shan Luo
- Department of Colorectal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Ru-Jing Lin
- Department of General Surgery, The People's Hospital of Binyang, Nanning, China
| | - Xi-Wen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun-Qiang Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
3
|
Satala D, Bednarek A, Kozik A, Rapala-Kozik M, Karkowska-Kuleta J. The Recruitment and Activation of Plasminogen by Bacteria-The Involvement in Chronic Infection Development. Int J Mol Sci 2023; 24:10436. [PMID: 37445613 DOI: 10.3390/ijms241310436] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
The development of infections caused by pathogenic bacteria is largely related to the specific properties of the bacterial cell surface and extracellular hydrolytic activity. Furthermore, a significant role of hijacking of host proteolytic cascades by pathogens during invasion should not be disregarded during consideration of the mechanisms of bacterial virulence. This is the key factor for the pathogen evasion of the host immune response, tissue damage, and pathogen invasiveness at secondary infection sites after initial penetration through tissue barriers. In this review, the mechanisms of bacterial impact on host plasminogen-the precursor of the important plasma serine proteinase, plasmin-are characterized, principally focusing on cell surface exposition of various proteins, responsible for binding of this host (pro)enzyme and its activators or inhibitors, as well as the fibrinolytic system activation tactics exploited by different bacterial species, not only pathogenic, but also selected harmless residents of the human microbiome. Additionally, the involvement of bacterial factors that modulate the process of plasminogen activation and fibrinolysis during periodontitis is also described, providing a remarkable example of a dual use of this host system in the development of chronic diseases.
Collapse
Affiliation(s)
- Dorota Satala
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Aneta Bednarek
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-387 Kraków, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| |
Collapse
|
4
|
Ali S, Karaynir A, Salih H, Öncü S, Bozdoğan B. Characterization, genome analysis and antibiofilm efficacy of lytic Proteus phages RP6 and RP7 isolated from university hospital sewage. Virus Res 2023; 326:199049. [PMID: 36717023 DOI: 10.1016/j.virusres.2023.199049] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/29/2023]
Abstract
The crystalline formation of biofilms by Proteus blocks the urine flow which often complicates the health care of catheterized patients. Bacteriophages has been highlighted as a promising tool to control biofilm-mediated bacterial infections. Here, we isolated and characterized two newly isolated lytic phages capable of infecting clinical isolates of P. mirabilis and P. vulgaris. Moreover, insights regarding the biological and molecular characterization were analysed. Both RP6 and RP7 phages showed a Proteus-genus-specific profile, administering no lytic activity against other family of Enterobacteriaceae. The optimal MOI value of the RP6 and RP7 phages were determined as 0.1 and 0.01, respectively. The one-step growth curve showed that RP6 and RP7 phages have a short latent period of 20 min and large burst size of 220-371 PFU/ML per infected host cell. Bacteria growth was reduced immediately after the phages were added, which is shown by the optical density (OD) measurement after 24 hr. Proteus phage RP6 and RP7 were found to eradicate both the planktonic and mature biofilms produced by the Proteus isolates tested. Genome sequence of Proteus phage RP6 was found to be 58,619 bp, and a G-C content of 47%. Also, Proteus phage RP7 genome size was 103,593 bp with G-C ratio of 38.45%. A total of 70 and 172 open reading frame (ORF) was encoded in RP6 and RP7 phage genomes, respectively. Interestingly, there were no tRNA encoded by Proteus phage RP6 genome even though there is a significant G-C content difference between the phage and its host. Additionally, the exhibition of highly lytic activity and absence of virulence and antibiotic-resistant genes in both Proteus RP6 and RP7 phages emphasized that this newly isolated phages are promising for potential therapeutic phages.
Collapse
Affiliation(s)
- Sahd Ali
- Recombinant DNA and Recombinant Protein Center (REDPROM), Aydın Adnan Menderes University, Turkiye.
| | - Abdulkerim Karaynir
- Recombinant DNA and Recombinant Protein Center (REDPROM), Aydın Adnan Menderes University, Turkiye
| | - Hanife Salih
- Recombinant DNA and Recombinant Protein Center (REDPROM), Aydın Adnan Menderes University, Turkiye
| | - Serkan Öncü
- Medical Faculty, Department of Infectious Diseases and Clinical Microbiology, Aydin Adnan Menderes University, Turkiye
| | - Bülent Bozdoğan
- Recombinant DNA and Recombinant Protein Center (REDPROM), Aydın Adnan Menderes University, Turkiye; Medical Faculty, Department of Medical Microbiology, Aydın Adnan Menderes University, Turkiye
| |
Collapse
|
5
|
Streptococcus mutans Proteases Degrade Dentinal Collagen. Dent J (Basel) 2022; 10:dj10120223. [PMID: 36547039 PMCID: PMC9776523 DOI: 10.3390/dj10120223] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Here, we explored the role of S. mutans’s whole cell and discrete fractions in the degradation of type I collagen and dentinal collagen. Type I collagen gels and human demineralized dentin slabs (DS) were incubated in media alone or with one of the following: overnight (O/N) or newly inoculated (NEW) cultures of S. mutans UA159; intracellular proteins, supernatant or bacterial membranes of O/N cultures. Media from all groups were analyzed for protease-mediated release of the collagen-specific imino acid hydroxyproline. Images of type I collagen and DS were analyzed, respectively. Type I collagen degradation was highest for the supernatant (p < 0.05) fractions, followed by intracellular components and O/N cultures. Collagen degradation for DS samples was highest for O/N samples, followed by supernatant, and intracellular components (p < 0.05). There was lower detectable degradation for both type I collagen and DS from NEW culture samples (p < 0.05), and there was no type I collagen or DS degradation detected for bacterial membrane samples. Structural changes to type I collagen gel and dentinal collagen were observed, respectively, following incubation with S. mutans cultures (O/N and NEW), intracellular components, and supernatant. This study demonstrates that intracellular and extracellular proteolytic activities from S. mutans enable this cariogenic bacterium to degrade type I and dentinal collagen in a growth-phase dependent manner, potentially contributing to the progression of dental caries.
Collapse
|
6
|
Harding A, Kanagasingam S, Welbury R, Singhrao SK. Periodontitis as a Risk Factor for Alzheimer's Disease: The Experimental Journey So Far, with Hope of Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1373:241-260. [PMID: 35612802 DOI: 10.1007/978-3-030-96881-6_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Periodontitis and Alzheimer's disease (AD) exist globally within the adult population. Given that the risk of AD incidence doubles within 10 years from the time of periodontal disease diagnosis, there is a window of opportunity for slowing down or preventing AD by risk-reduction-based intervention. Literature appraisal on the shared risk factors of these diseases suggests a shift to a healthy lifestyle would be beneficial. Generalised (chronic) periodontitis with an established dysbiotic polymicrobial aetiology affects the tooth supporting tissues with eventual tooth loss. The cause of AD remains unknown, however two neurohistopathological lesions - amyloid-beta plaques and neurofibrillary tangles, together with the clinical history, provide AD diagnosis at autopsy. Historically, prominence was given to the two hallmark lesions but now emphasis is placed on cerebral inflammation and what triggers it. Low socioeconomic status promotes poor lifestyles that compromise oral and personal hygiene along with reliance on poor dietary intake. Taken together with advancing age and a declining immune protection, these risk factors may negatively impact on periodontitis and AD. These factors also provide a tangible solution to controlling pathogenic bacteria indigenous to the oral and gastrointestinal tract microbioes in vulnerable subjects. The focus here is on Porphyromonas gingivalis, one of several important bacterial pathogens associated with both periodontitis and AD. Recent research has enabled advances in our knowledge of the armoury of P. gingivalis via reproduction of all clinical and neuropathological hallmark lesions of AD and chronic periodontal disease in vitro and in vivo experimental models, thus paving the way for better future management.
Collapse
Affiliation(s)
- Alice Harding
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Shalini Kanagasingam
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Richard Welbury
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Sim K Singhrao
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK.
| |
Collapse
|
7
|
Wadhawan A, Reynolds MA, Makkar H, Scott AJ, Potocki E, Hoisington AJ, Brenner LA, Dagdag A, Lowry CA, Dwivedi Y, Postolache TT. Periodontal Pathogens and Neuropsychiatric Health. Curr Top Med Chem 2021; 20:1353-1397. [PMID: 31924157 DOI: 10.2174/1568026620666200110161105] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 02/08/2023]
Abstract
Increasing evidence incriminates low-grade inflammation in cardiovascular, metabolic diseases, and neuropsychiatric clinical conditions, all important causes of morbidity and mortality. One of the upstream and modifiable precipitants and perpetrators of inflammation is chronic periodontitis, a polymicrobial infection with Porphyromonas gingivalis (P. gingivalis) playing a central role in the disease pathogenesis. We review the association between P. gingivalis and cardiovascular, metabolic, and neuropsychiatric illness, and the molecular mechanisms potentially implicated in immune upregulation as well as downregulation induced by the pathogen. In addition to inflammation, translocation of the pathogens to the coronary and peripheral arteries, including brain vasculature, and gut and liver vasculature has important pathophysiological consequences. Distant effects via translocation rely on virulence factors of P. gingivalis such as gingipains, on its synergistic interactions with other pathogens, and on its capability to manipulate the immune system via several mechanisms, including its capacity to induce production of immune-downregulating micro-RNAs. Possible targets for intervention and drug development to manage distal consequences of infection with P. gingivalis are also reviewed.
Collapse
Affiliation(s)
- Abhishek Wadhawan
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States.,Department of Psychiatry, Saint Elizabeths Hospital, Washington, D.C. 20032, United States
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore 21201, United States
| | - Hina Makkar
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States
| | - Alison J Scott
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, United States
| | - Eileen Potocki
- VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, United States
| | - Andrew J Hoisington
- Air Force Institute of Technology, Wright-Patterson Air Force Base, United States
| | - Lisa A Brenner
- Departments of Psychiatry, Neurology, and Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, United States.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, United States
| | - Aline Dagdag
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States
| | - Christopher A Lowry
- Departments of Psychiatry, Neurology, and Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, United States.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, United States.,Department of Integrative Physiology, Center for Neuroscience and Center for Microbial Exploration, University of Colorado Boulder, Boulder, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, United States
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Alabama, United States
| | - Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, United States.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, United States.,Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, United States
| |
Collapse
|
8
|
Hasan R, Rony MNH, Ahmed R. In silico characterization and structural modeling of bacterial metalloprotease of family M4. J Genet Eng Biotechnol 2021; 19:25. [PMID: 33528696 PMCID: PMC7851659 DOI: 10.1186/s43141-020-00105-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 12/15/2020] [Indexed: 01/20/2023]
Abstract
BACKGROUND The M4 family of metalloproteases is comprised of a large number of zinc-containing metalloproteases. A large number of these enzymes are important virulence factors of pathogenic bacteria and therefore potential drug targets. Whereas some enzymes have potential for biotechnological applications, the M4 family of metalloproteases is known almost exclusively from bacteria. The aim of the study was to identify the structure and properties of M4 metalloprotease proteins. RESULTS A total of 31 protein sequences of M4 metalloprotease retrieved from UniProt representing different species of bacteria have been characterized for various physiochemical properties. They were thermostable, hydrophillic protein of a molecular mass ranging from 38 to 66 KDa. Correlation on the basis of both enzymes and respective genes has also been studied by phylogenetic tree. B. cereus M4 metalloprotease (PDB ID: 1NPC) was selected as a representative species for secondary and tertiary structures among the M4 metalloprotease proteins. The secondary structure displaying 11 helices (H1-H11) is involved in 15 helix-helix interactions, while 4 β-sheet motifs composed of 15 β-strands in PDBsum. Possible disulfide bridges were absent in most of the cases. The tertiary structure of B. cereus M4 metalloprotease was validated by QMEAN4 and SAVES server (Ramachandran plot, verify 3D, and ERRAT) which proved the stability, reliability, and consistency of the tertiary structure of the protein. Functional analysis was done in terms of membrane protein topology, disease-causing region prediction, proteolytic cleavage sites prediction, and network generation. Transmembrane helix prediction showed absence of transmembrane helix in protein. Protein-protein interaction networks demonstrated that bacillolysin of B. cereus interacted with ten other proteins in a high confidence score. Five disorder regions were identified. Active sites analysis showed the zinc-binding residues-His-143, His-147, and Glu-167, with Glu-144 acting as the catalytic residues. CONCLUSION Moreover, this theoretical overview will help researchers to get a details idea about the protein structure and it may also help to design enzymes with desirable characteristics for exploiting them at industrial level or potential drug targets.
Collapse
Affiliation(s)
- Rajnee Hasan
- Basic and Applied Research on Jute Project, Bangladesh Jute Research Institute, Manik Mia Avenue, Dhaka, 1207 Bangladesh
| | - Md. Nazmul Haq Rony
- Basic and Applied Research on Jute Project, Bangladesh Jute Research Institute, Manik Mia Avenue, Dhaka, 1207 Bangladesh
| | - Rasel Ahmed
- Basic and Applied Research on Jute Project, Bangladesh Jute Research Institute, Manik Mia Avenue, Dhaka, 1207 Bangladesh
| |
Collapse
|
9
|
Sandri A, Lleo MM, Signoretto C, Boaretti M, Boschi F. Protease inhibitors elicit anti-inflammatory effects in CF mice with Pseudomonas aeruginosa acute lung infection. Clin Exp Immunol 2020; 203:87-95. [PMID: 32946591 DOI: 10.1111/cei.13518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/03/2020] [Accepted: 09/09/2020] [Indexed: 01/30/2023] Open
Abstract
Pseudomonas aeruginosa is the major respiratory pathogen in patients with cystic fibrosis (CF). P. aeruginosa-secreted proteases, in addition to host proteases, degrade lung tissue and interfere with immune processes. In this study, we aimed at evaluating the possible anti-inflammatory effects of protease inhibitors Marimastat and Ilomastat in the treatment of P. aeruginosa infection. Lung infection with the P. aeruginosa PAO1 strain was established in wild-type and cystic fibrosis transmembrane conductance regulator (CFTR) knock-out C57BL/6 mice expressing a luciferase gene under control of bovine interleukin (IL)-8 promoter. After intratracheal instillation with 150 µM Marimastat and Ilomastat, lung inflammation was monitored by in-vivo bioluminescence imaging and bacterial load in the lungs was assessed. In vitro, the effects of protease inhibitors on PAO1 growth and viability were evaluated. Acute lung infection was established in both wild-type and CFTR knock-out mice. After 24 h, the infection induced IL-8-dependent bioluminescence emission, indicating lung inflammation. In infected mice with ongoing inflammation, intratracheal treatment with 150 µM Marimastat and Ilomastat reduced the bioluminescence signal in comparison to untreated, infected animals. Bacterial load in the lungs was not affected by the treatment, and in vitro the same dose of Marimastat and Ilomastat did not affect PAO1 growth and viability, confirming that these molecules have no additional anti-bacterial activity. Our results show that inhibition of protease activity elicits anti-inflammatory effects in cystic fibrosis (CF) mice with acute P. aeruginosa lung infection. Thus, Marimastat and Ilomastat represent candidate molecules for the treatment of CF patients, encouraging further studies on protease inhibitors and their application in inflammatory diseases.
Collapse
Affiliation(s)
- A Sandri
- Department of Diagnostics and Public Health, Section of Microbiology, University of Verona, Verona, Italy
| | - M M Lleo
- Department of Diagnostics and Public Health, Section of Microbiology, University of Verona, Verona, Italy
| | - C Signoretto
- Department of Diagnostics and Public Health, Section of Microbiology, University of Verona, Verona, Italy
| | - M Boaretti
- Department of Diagnostics and Public Health, Section of Microbiology, University of Verona, Verona, Italy
| | - F Boschi
- Department of Computer Science, University of Verona, Verona, Italy
| |
Collapse
|
10
|
Experimental and theoretical studies of imidazole based chemosensor for Palladium and their biological applications. J Photochem Photobiol A Chem 2019. [DOI: 10.1016/j.jphotochem.2019.112092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Yang C, Yu T, Liu Z, Ye X, Liao X, Wang X, Han C, Zhu G, Qin W, Peng T. Cystatin F as a key family 2 cystatin subunit and prognostic biomarker for early‑stage pancreatic ductal adenocarcinoma. Oncol Rep 2019; 42:79-90. [PMID: 31059105 PMCID: PMC6549077 DOI: 10.3892/or.2019.7135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 04/17/2019] [Indexed: 01/03/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, and early diagnosis and assessment may enhance the quality of life and survival of patients. The prognostic value of key family 2 cystatins subunit in PDAC patients remains unknown. The potential molecular roles of family 2 cystatins and related pathways were investigated using bioinformatics analysis. The relationship of family 2 cystatin expression levels and clinical outcomes of 112 patients with early-stage PDAC were evaluated via univariate and combined survival analysis. A prognostic nomogram model was also constructed and gene set enrichment analysis was performed to investigate potential pathways in PDAC. The pathways, interaction networks, and Gene Ontology term analysis of the cystatin gene family were analyzed in the present study. Cystatin F (CST7) was identified as the key subunit of family 2 cystatins in survival analysis. PDAC patients who harbored a higher expression level of CST7 had a lower risk in overall survival (adjusted HROS=0.44, 95% CI=0.25-0.77, P=0.004) and a longer survival time in various subgroups. The prognostic nomogram indicated that the CST7 expression model effectively predicted the outcomes of patients with early-stage PDAC (predictive ability >0.75). In the gene set enrichment analysis, it was revealed that CST7 expression may be involved in immune regulation and be associated with cell adhesion. CST7 could be a useful biomarker for the prognostic prediction of early-stage PDAC after pancreaticoduodenectomy.
Collapse
Affiliation(s)
- Chengkun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Tingdong Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhengqian Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Wei Qin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
12
|
Minasyan H, Flachsbart F. Blood coagulation: a powerful bactericidal mechanism of human innate immunity. Int Rev Immunol 2019; 38:3-17. [DOI: 10.1080/08830185.2018.1533009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hayk Minasyan
- Private laboratory, Immunology Microbiology, Yerevan, Armenia
| | | |
Collapse
|
13
|
Dominy SS, Lynch C, Ermini F, Benedyk M, Marczyk A, Konradi A, Nguyen M, Haditsch U, Raha D, Griffin C, Holsinger LJ, Arastu-Kapur S, Kaba S, Lee A, Ryder MI, Potempa B, Mydel P, Hellvard A, Adamowicz K, Hasturk H, Walker GD, Reynolds EC, Faull RLM, Curtis MA, Dragunow M, Potempa J. Porphyromonas gingivalis in Alzheimer's disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. SCIENCE ADVANCES 2019; 5:eaau3333. [PMID: 30746447 PMCID: PMC6357742 DOI: 10.1126/sciadv.aau3333] [Citation(s) in RCA: 1059] [Impact Index Per Article: 211.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 12/11/2018] [Indexed: 05/19/2023]
Abstract
Porphyromonas gingivalis, the keystone pathogen in chronic periodontitis, was identified in the brain of Alzheimer's disease patients. Toxic proteases from the bacterium called gingipains were also identified in the brain of Alzheimer's patients, and levels correlated with tau and ubiquitin pathology. Oral P. gingivalis infection in mice resulted in brain colonization and increased production of Aβ1-42, a component of amyloid plaques. Further, gingipains were neurotoxic in vivo and in vitro, exerting detrimental effects on tau, a protein needed for normal neuronal function. To block this neurotoxicity, we designed and synthesized small-molecule inhibitors targeting gingipains. Gingipain inhibition reduced the bacterial load of an established P. gingivalis brain infection, blocked Aβ1-42 production, reduced neuroinflammation, and rescued neurons in the hippocampus. These data suggest that gingipain inhibitors could be valuable for treating P. gingivalis brain colonization and neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Stephen S. Dominy
- Cortexyme, Inc., 269 East Grand Ave., South San Francisco, CA, USA
- Corresponding author.
| | - Casey Lynch
- Cortexyme, Inc., 269 East Grand Ave., South San Francisco, CA, USA
| | - Florian Ermini
- Cortexyme, Inc., 269 East Grand Ave., South San Francisco, CA, USA
| | - Malgorzata Benedyk
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Agata Marczyk
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Andrei Konradi
- Cortexyme, Inc., 269 East Grand Ave., South San Francisco, CA, USA
| | - Mai Nguyen
- Cortexyme, Inc., 269 East Grand Ave., South San Francisco, CA, USA
| | - Ursula Haditsch
- Cortexyme, Inc., 269 East Grand Ave., South San Francisco, CA, USA
| | - Debasish Raha
- Cortexyme, Inc., 269 East Grand Ave., South San Francisco, CA, USA
| | | | | | | | - Samer Kaba
- Cortexyme, Inc., 269 East Grand Ave., South San Francisco, CA, USA
| | - Alexander Lee
- Cortexyme, Inc., 269 East Grand Ave., South San Francisco, CA, USA
| | - Mark I. Ryder
- Division of Periodontology, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Barbara Potempa
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Piotr Mydel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Broegelman Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Annelie Hellvard
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Broegelman Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Karina Adamowicz
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Hatice Hasturk
- The Forsyth Institute, Cambridge, MA, USA
- Harvard University School of Dental Medicine, Boston, MA, USA
| | - Glenn D. Walker
- Cooperative Research Centre for Oral Health Science, Melbourne Dental School and the Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Victoria, Australia
| | - Eric C. Reynolds
- Cooperative Research Centre for Oral Health Science, Melbourne Dental School and the Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Victoria, Australia
| | - Richard L. M. Faull
- Department of Anatomy with Radiology, Centre for Brain Research and NeuroValida, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Maurice A. Curtis
- Centre for Brain Research and NeuroValida, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Centre for Brain Research and NeuroValida, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| |
Collapse
|
14
|
Eisenhardt M, Schlupp P, Höfer F, Schmidts T, Hoffmann D, Czermak P, Pöppel AK, Vilcinskas A, Runkel F. The therapeutic potential of the insect metalloproteinase inhibitor against infections caused by Pseudomonas aeruginosa. J Pharm Pharmacol 2018; 71:316-328. [PMID: 30408181 DOI: 10.1111/jphp.13034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/29/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The objective of this study was to investigate the therapeutic potential of the insect metalloproteinase inhibitor (IMPI) from Galleria mellonella, the only known specific inhibitor of M4 metalloproteinases. METHODS The fusion protein IMPI-GST (glutathione-S-transferase) was produced by fermentation in Escherichia coli and was tested for its ability to inhibit the proteolytic activity of the M4 metalloproteinases thermolysin and Pseudomonas elastase (PE), the latter a key virulence factor of the wound-associated and antibiotic-resistant pathogen Pseudomonas aeruginosa. We also tested the ability of IMPI to inhibit the secretome (Sec) of a P. aeruginosa strain obtained from a wound. KEY FINDINGS We found that IMPI-GST inhibited thermolysin and PE in vitro and increased the viability of human keratinocytes exposed to Sec by inhibiting detachment caused by changes in cytoskeletal morphology. IMPI-GST also improved the cell migration rate in an in vitro wound assay and reduced the severity of necrosis caused by Sec in an ex vivo porcine wound model. CONCLUSIONS The inhibition of virulence factors is a novel therapeutic approach against antibiotic resistant bacteria. Our results indicate that IMPI is a promising drug candidate for the treatment of P. aeruginosa infections.
Collapse
Affiliation(s)
- Michaela Eisenhardt
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Technische Hochschule Mittelhessen, Giessen, Germany
| | - Peggy Schlupp
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Technische Hochschule Mittelhessen, Giessen, Germany
| | - Frank Höfer
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Technische Hochschule Mittelhessen, Giessen, Germany
| | - Thomas Schmidts
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Technische Hochschule Mittelhessen, Giessen, Germany
| | - Daniel Hoffmann
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Technische Hochschule Mittelhessen, Giessen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Technische Hochschule Mittelhessen, Giessen, Germany.,Department of Bio-Resources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Giessen, Germany
| | - Anne-Kathrin Pöppel
- Department of Bio-Resources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Giessen, Germany
| | - Andreas Vilcinskas
- Department of Bio-Resources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Giessen, Germany.,Institute for Insect Biotechnology, Justus Liebig University of Giessen, Giessen, Germany
| | - Frank Runkel
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Technische Hochschule Mittelhessen, Giessen, Germany
| |
Collapse
|
15
|
Safety Aspect of Enterococcus faecium FL31 Strain and Antibacterial Mechanism of Its Hydroxylated Bacteriocin BacFL31 against Listeria monocytogenes. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5308464. [PMID: 30515405 PMCID: PMC6236939 DOI: 10.1155/2018/5308464] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/09/2018] [Indexed: 11/17/2022]
Abstract
In previous work we have isolated and identified a new strain called Enterococcus faecium FL31. The active compound secreted by this strain, "BacFL31", has been purified and characterized. In the present study, safety aspect, assessed by microbiological and molecular tests, demonstrated that Enterococcus faecium FL31 was susceptible to relevant antibiotics, free of hemolytic, gelatinase, DNase, and lipase activities. In addition, it did not harbor virulence and antibiotic resistance genes. Combined SYTOX Green dye and UV-absorbing experiments, along with released extracellular potassium and transmembrane electrical potential measurements, showed that pure BacFL31 at a concentration of 1×MIC (50 μg/mL) could damage cytoplasmic membrane of the pathogen Listeria monocytogenes ATCC19117. The same concentration causes the leakage of its intracellular constituents and leads to the destruction of this pathogenic microorganism. In summary, this work reflected characteristics of Enterococcus faecium FL31 strain and its bacteriocin in terms of functional and safety perspectives.
Collapse
|
16
|
Sandri A, Ortombina A, Boschi F, Cremonini E, Boaretti M, Sorio C, Melotti P, Bergamini G, Lleo M. Inhibition of Pseudomonas aeruginosa secreted virulence factors reduces lung inflammation in CF mice. Virulence 2018; 9:1008-1018. [PMID: 29938577 PMCID: PMC6086295 DOI: 10.1080/21505594.2018.1489198] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background: Cystic fibrosis (CF) lung infection is a complex condition where opportunistic pathogens and defective immune system cooperate in developing a constant cycle of infection and inflammation. The major pathogen, Pseudomonas aeruginosa, secretes a multitude of virulence factors involved in host immune response and lung tissue damage. In this study, we examined the possible anti-inflammatory effects of molecules inhibiting P. aeruginosa virulence factors. Methods: Pyocyanin, pyoverdine and proteases were measured in bacterial culture supernatant from different P. aeruginosa strains. Inhibition of virulence factors by sub-inhibitory concentrations of clarithromycin and by protease inhibitors was evaluated. Lung inflammatory response was monitored by in vivo bioluminescence imaging in wild-type and CFTR-knockout mice expressing a luciferase gene under the control of a bovine IL-8 promoter. Results: The amount of proteases, pyocyanin and pyoverdine secreted by P. aeruginosa strains was reduced after growth in the presence of a sub-inhibitory dose of clarithromycin. Intratracheal challenge with culture supernatant containing bacteria-released products induced a strong IL-8-mediated response in mouse lungs while lack of virulence factors corresponded to a reduction in bioluminescence emission. Particularly, sole inactivation of proteases by inhibitors Ilomastat and Marimastat also resulted in decreased lung inflammation. Conclusions: Our data support the assumption that virulence factors are involved in P. aeruginosa pro-inflammatory action in CF lungs; particularly, proteases seem to play an important role. Inhibition of virulence factors production and activity resulted in decreased lung inflammation; thus, clarithromycin and protease inhibitors potentially represent additional therapeutic therapies for P. aeruginosa-infected patients.
Collapse
Affiliation(s)
- Angela Sandri
- a Department of Diagnostics and Public Health , University of Verona , Verona , Italy
| | - Alessia Ortombina
- a Department of Diagnostics and Public Health , University of Verona , Verona , Italy
| | - Federico Boschi
- b Department of Computer Science , University of Verona , Verona , Italy
| | - Eleonora Cremonini
- a Department of Diagnostics and Public Health , University of Verona , Verona , Italy
| | - Marzia Boaretti
- a Department of Diagnostics and Public Health , University of Verona , Verona , Italy
| | - Claudio Sorio
- c Department of Medicine , University of Verona , Verona , Italy
| | - Paola Melotti
- d Cystic Fibrosis Center , Azienda Ospedaliera Universitaria Integrata di Verona , Verona , Italy
| | | | - Maria Lleo
- a Department of Diagnostics and Public Health , University of Verona , Verona , Italy
| |
Collapse
|
17
|
Kany AM, Sikandar A, Yahiaoui S, Haupenthal J, Walter I, Empting M, Köhnke J, Hartmann RW. Tackling Pseudomonas aeruginosa Virulence by a Hydroxamic Acid-Based LasB Inhibitor. ACS Chem Biol 2018; 13:2449-2455. [PMID: 30088919 DOI: 10.1021/acschembio.8b00257] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In search of novel antibiotics to combat the challenging spread of resistant pathogens, bacterial proteases represent promising targets for pathoblocker development. A common motif for protease inhibitors is the hydroxamic acid function, yet this group has often been related to unspecific inhibition of various metalloproteases. In this work, the inhibition of LasB, a harmful zinc metalloprotease secreted by Pseudomonas aeruginosa, through a hydroxamate derivative is described. The present inhibitor was developed based on a recently reported, highly selective thiol scaffold. Using X-ray crystallography, the lack of inhibition of a range of human matrix metalloproteases could be attributed to a distinct binding mode sparing the S1' pocket. The inhibitor was shown to restore the effect of the antimicrobial peptide LL-37, decrease the formation of P. aeruginosa biofilm and, for the first time for a LasB inhibitor, reduce the release of extracellular DNA. Hence, it is capable of disrupting several important bacterial resistance mechanisms. These results highlight the potential of protease inhibitors to fight bacterial infections and point out the possibility to achieve selective inhibition even with a strong zinc anchor.
Collapse
Affiliation(s)
- Andreas M. Kany
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Asfandyar Sikandar
- Workgroup Structural Biology of Biosynthetic Enzymes, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Samir Yahiaoui
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Jörg Haupenthal
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Isabell Walter
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Martin Empting
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Jesko Köhnke
- Workgroup Structural Biology of Biosynthetic Enzymes, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Rolf W. Hartmann
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
18
|
Amala S, Santhi S, Subashini S. Synthesis, Characterization, Analytical and Cytotoxic Studies of N,N′-Bis(2-Hydroxynaphthylidene)-4-chlorophenyl Methanediamine and Its Co(II), Ni(II) and Cu(II) Complexes. ChemistrySelect 2018. [DOI: 10.1002/slct.201801406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Subbiah Amala
- PG and Research Department of Chemistry; Seethalakshmi Ramaswami College; Tiruchirappalli 620 002 Tamil Nadu India
| | - Sambamoorthy Santhi
- PG and Research Department of Chemistry; Seethalakshmi Ramaswami College; Tiruchirappalli 620 002 Tamil Nadu India
| | - Sungapillai Subashini
- PG and Research Department of Chemistry; Seethalakshmi Ramaswami College; Tiruchirappalli 620 002 Tamil Nadu India
| |
Collapse
|
19
|
Jiang J, Liu HL, Tao L, Lin XY, Yang YD, Tan SW, Wu B. Let‑7d inhibits colorectal cancer cell proliferation through the CST1/p65 pathway. Int J Oncol 2018; 53:781-790. [PMID: 29845224 DOI: 10.3892/ijo.2018.4419] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 04/19/2018] [Indexed: 12/14/2022] Open
Abstract
Cystatin SN (cystatin 1, CST1) is a member of the cystatin superfamily which inhibits the proteolytic activity of cysteine proteases. CST1 is a tumor biomarker that provides useful information for the diagnosis of esophageal, gastric and colorectal carcinomas. MicroRNAs (miRNAs or miRs) play an important role in tumor cell proliferation. However, the exact role of let‑7d and CST1 in colon cancer remains unknown. The aim of this study was to assess whether let‑7d inhibits colorectal carcinogenesis through the CST1/p65 pathway, and determine whether it may be used as a potential target for clinical therapy. Microarray analysis of mRNAs extracted from colon cancer and normal tissues was performed. The results of gene expression microanalysis revealed that CST1 expression was upregulated in colon cancer compared with normal tissues. In addition, the upregulation of CST1 expression and the downregulation of let‑7d expression in patients with colon cancer and in several colorectal cancer cell lines were confirmed by reverse transcription-quantitative PCR (RT‑qPCR), immunohistochemistry and western blot analysis. In addition, siRNA targeting CST1 (CST1‑siRNA) and let‑7d-mimics were used in the HCT116 cells, and the results revealed that CST1 and let‑7d played a role in colorectal cancer cell proliferation. Let‑7d inhibited colorectal carcinogenesis through the CST1/p65 pathway. Thus, the findings of the present study indicate that CST1 may be a potential target for the future clinical therapy of colorectal cancer.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Hui-Ling Liu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Li Tao
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Xian-Yi Lin
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Yi-Dong Yang
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Si-Wei Tan
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Bin Wu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
20
|
Rovai ES, Holzhausen M. The Role of Proteinase-Activated Receptors 1 and 2 in the Regulation of Periodontal Tissue Metabolism and Disease. J Immunol Res 2017; 2017:5193572. [PMID: 28503577 PMCID: PMC5414592 DOI: 10.1155/2017/5193572] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/13/2017] [Accepted: 03/05/2017] [Indexed: 01/13/2023] Open
Abstract
Proteinase-activated receptors 1 (PAR1) and 2 (PAR2) are the most highly expressed members of the PAR family in the periodontium. These receptors regulate periodontal inflammatory and repair processes through their activation by endogenous and bacterial enzymes. PAR1 is expressed by the periodontal cells such as human gingival fibroblasts, gingival epithelial cells, periodontal ligament cells, osteoblasts, and monocytic cells and can be activated by thrombin, matrix metalloproteinase 1 (MMP-1), MMP-13, fibrin, and gingipains from Porphyromonas gingivalis. PAR2 is expressed by neutrophils, osteoblasts, oral epithelial cells, and human gingival fibroblasts, and its possible activators in the periodontium are gingipains, neutrophil proteinase 3, and mast cell tryptase. The mechanisms through which PARs can respond to periodontal enzymes and result in appropriate immune responses have until recently been poorly understood. This review discusses recent findings that are beginning to identify a cardinal role for PAR1 and PAR2 on periodontal tissue metabolism.
Collapse
MESH Headings
- Adhesins, Bacterial/metabolism
- Animals
- Cells, Cultured
- Cysteine Endopeptidases/metabolism
- Epithelial Cells
- Fibroblasts
- Gene Expression Regulation
- Gingipain Cysteine Endopeptidases
- Gingiva/cytology
- Gingiva/metabolism
- Humans
- Matrix Metalloproteinase 1/genetics
- Matrix Metalloproteinase 1/metabolism
- Mice
- Periodontitis/genetics
- Periodontitis/metabolism
- Periodontitis/physiopathology
- Periodontium/metabolism
- Porphyromonas gingivalis
- Receptor, PAR-1/agonists
- Receptor, PAR-1/antagonists & inhibitors
- Receptor, PAR-1/genetics
- Receptor, PAR-1/metabolism
- Receptors, Proteinase-Activated/agonists
- Receptors, Proteinase-Activated/antagonists & inhibitors
- Receptors, Proteinase-Activated/genetics
- Receptors, Proteinase-Activated/metabolism
Collapse
Affiliation(s)
- E. S. Rovai
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - M. Holzhausen
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
21
|
Taniguchi M, Matsuhashi Y, Abe TK, Ishiyama Y, Saitoh E, Kato T, Ochiai A, Tanaka T. Contribution of cationic amino acids toward the inhibition of Arg-specific cysteine proteinase (Arg-gingipain) by the antimicrobial dodecapeptide, CL(14-25), from rice protein. Biopolymers 2016; 102:379-89. [PMID: 25046435 DOI: 10.1002/bip.22525] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 06/11/2014] [Accepted: 06/28/2014] [Indexed: 11/11/2022]
Abstract
CL(14-25), a dodecapeptide, exhibits antimicrobial activity against Porphyromonas gingivalis with the 50% growth-inhibitory concentration (IC50 ) value of 145 µM, and arginine-specific gingipain (Rgp)-inhibitory activity. Kinetic analysis revealed that CL(14-25) is a mixed-type inhibitor, with inhibition constants (Ki and Ki ' values) of 1.4 × 10(-6) M and 4.3 × 10(-6) M, respectively. To elucidate the contributions of four cationic amino acid residues at the N- and C-termini of CL(14-25) toward Rgp-inhibitory activity, we investigated the Rgp-inhibitory activities of truncated and alanine-substituted analogs of CL(14-25). Rgp-inhibitory activities significantly decreased by truncated analogs, CL(15-25) and CL(16-25), whereas those of CL(14-24) and CL(14-23) were almost as high as that of CL(14-25). Rgp-inhibitory activities of alanine-substituted analogs, CL(R14A) and CL(R14A, R15A) also significantly decreased, whereas those of CL(K25A) and CL(R24A, K25A) were higher than that of CL(14-25). These results suggest that the arginine residue at position 15 substantially contributes to the Rgp-inhibitory activity and that the arginine residue at position 14 plays important roles in exerting Rgp-inhibitory activity. In this study, we demonstrated that CL(K25A) was a potent, dual function, peptide inhibitor candidate, exhibiting Rgp-inhibitory activity with Ki and Ki ' of 9.6 × 10(-7) M and 1.9 × 10(-6) M, respectively, and antimicrobial activity against P. gingivalis with an IC50 value of 51 µM.
Collapse
Affiliation(s)
- Masayuki Taniguchi
- Department of Materials Science and Technology, Graduate School of Science and Technology, Niigata University, Niigata, 950-2181, Japan; Center for Transdisciplinary Research, Niigata University, Niigata, 950-2181, Japan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Dabhade AR, Mokashe NU, Patil UK. Purification, characterization, and antimicrobial activity of nontoxic trypsin inhibitor from Albizia amara Boiv. Process Biochem 2016. [DOI: 10.1016/j.procbio.2016.02.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
23
|
Jordan RE, Fernandez J, Brezski RJ, Greenplate AR, Knight DM, Raju TS, Lynch AS. A peptide immunization approach to counteract a Staphylococcus aureus protease defense against host immunity. Immunol Lett 2016; 172:29-39. [PMID: 26905931 DOI: 10.1016/j.imlet.2016.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 02/05/2016] [Accepted: 02/16/2016] [Indexed: 01/01/2023]
Abstract
Pathogens that induce acute and chronic infections, as well as certain cancers, employ numerous strategies to thwart host cellular and humoral immune defenses. One proposed evasion mechanism against humoral immunity is a localized expression of extracellular proteases that cleave the IgG hinge and disable host IgG functions. Host immunity appears to be prepared to counter such a proteolytic tactic by providing a group of autoantibodies, denoted anti-hinge antibodies that specifically bind to cleaved IgGs and provide compensating functional restoration in vitro. These respective counter-measures highlight the complex interrelationships among pathogens and host immunity and suggested to us a possible means for therapeutic intervention. In this study, we combined an investigation of pathogen-mediated proteolysis of host IgGs with an immunization strategy to boost host anti-hinge antibodies. In a Staphylococcus aureus infection model using an artificial tissue cage (wiffle ball) implanted into rabbits, cleaved rabbit IgGs were detected in abundance in the abscesses of untreated animals early after infection. However, in animals previously immunized with peptide analogs of the cleaved IgG hinge to generate substantial anti-hinge antibody titers, S. aureus colony formation was markedly reduced compared to control animals or those similarly immunized with a scrambled peptide sequence. The results of this study demonstrate that extensive local proteolysis of IgGs occurs in a test abscess setting and that immunization to increase host anti-hinge antibodies provided substantial acute protection against bacterial growth.
Collapse
Affiliation(s)
- Robert E Jordan
- Biologics Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477 USA.
| | - Jeffrey Fernandez
- Infectious Diseases and Vaccines, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA.
| | - Randall J Brezski
- Biologics Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477 USA
| | - Allison R Greenplate
- Biologics Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477 USA
| | - David M Knight
- Biologics Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477 USA
| | - T Shantha Raju
- Biologics Research, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477 USA
| | - A Simon Lynch
- Infectious Diseases and Vaccines, Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA 19477, USA
| |
Collapse
|
24
|
Abstract
As our knowledge of host-microbial interactions within the oral cavity increases, future treatments are likely to be more targeted. For example, efforts to target a single species or key virulence factors that they produce, while maintaining the natural balance of the resident oral microbiota that acts to modulate the host immune response would be an advantage. Targeted approaches may be directed at the black-pigmented anaerobes, Porphyromonas gingivalis and Prevotella intermedia, associated with periodontitis. Such pigments provide an opportunity for targeted phototherapy with high-intensity monochromatic light. Functional inhibition approaches, including the use of enzyme inhibitors, are also being explored to control periodontitis. More general disruption of dental plaque through the use of enzymes and detergents, alone and in combination, shows much promise. The use of probiotics and prebiotics to improve gastrointestinal health has now led to an interest in using these approaches to control oral disease. More recently the potential of antimicrobial peptides and nanotechnology, through the application of nanoparticles with biocidal, anti-adhesive and delivery capabilities, has been explored. The aim of this review is to consider the current status as regards non-conventional treatment approaches for oral infections with particular emphasis on the plaque-related diseases.
Collapse
Affiliation(s)
- Robert P Allaker
- a Oral Microbiology; Barts and The London School of Medicine & Dentistry; Queen Mary University of London ; London, UK
| | | |
Collapse
|
25
|
Inhibition of in vitro adhesion and virulence of Porphyromonas gingivalis by aqueous extract and polysaccharides from Rhododendron ferrugineum L. A new way for prophylaxis of periodontitis? Fitoterapia 2015; 107:105-113. [DOI: 10.1016/j.fitote.2015.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 12/30/2022]
|
26
|
Sjøli S, Nuti E, Camodeca C, Bilto I, Rossello A, Winberg JO, Sylte I, Adekoya OA. Synthesis, experimental evaluation and molecular modelling of hydroxamate derivatives as zinc metalloproteinase inhibitors. Eur J Med Chem 2015; 108:141-153. [PMID: 26638045 DOI: 10.1016/j.ejmech.2015.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/09/2015] [Accepted: 11/13/2015] [Indexed: 10/22/2022]
Abstract
Enzymes of the M4 family of zinc-metalloproteinases are virulence factors secreted from gram-positive or gram-negative bacteria, and putative drug targets in the treatment of bacterial infections. In order to have a therapeutic value such inhibitors should not interfere with endogenous zinc-metalloproteinases. In the present study we have synthesised a series of hydroxamate derivatives and validated the compounds as inhibitors of the M4 enzymes thermolysin and pseudolysin, and the endogenous metalloproteinases ADAM-17, MMP-2 and MMP-9 using experimental binding studies and molecular modelling. In general, the compounds are stronger inhibitors of the MMPs than of the M4 enzymes, however, an interesting exception is LM2. The compounds bound stronger to pseudolysin than to thermolysin, and the molecular modelling studies showed that occupation of the S2(') subpocket by an aromatic group is favourable for strong interactions with pseudolysin.
Collapse
Affiliation(s)
- Stian Sjøli
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, NO-9037 Tromsø, Norway
| | - Elisa Nuti
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Caterina Camodeca
- Division of Immunology, Transplants and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Irina Bilto
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, NO-9037 Tromsø, Norway
| | - Armando Rossello
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Jan-Olof Winberg
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, NO-9037 Tromsø, Norway
| | - Ingebrigt Sylte
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | - Olayiwola A Adekoya
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, NO-9037 Tromsø, Norway
| |
Collapse
|
27
|
Tetradentate hydrazone metal complexes derived from cefazolin and 2,6-diacetylpyridine hydrazide: synthesis, characterization, and antibacterial activity. MONATSHEFTE FUR CHEMIE 2015. [DOI: 10.1007/s00706-015-1585-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
28
|
Anacona JR, Rincones M. Tridentate hydrazone metal complexes derived from cephalexin and 2-hydrazinopyridine: synthesis, characterization and antibacterial activity. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2015; 141:169-175. [PMID: 25677531 DOI: 10.1016/j.saa.2015.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 11/17/2014] [Accepted: 01/05/2015] [Indexed: 06/04/2023]
Abstract
Metal(II) coordination compounds of a tridentate hydrazone ligand (HL) derived from the condensation of cephalexin antibiotic with 2-hydrazinopyridine were synthesized. The hydrazone ligand and mononuclear [ML(OAc)(H2O)] (M(II)=Mn, Co, Ni, Cu, Zn, Ag) complexes were characterized by several techniques, including elemental and thermal analysis, molar conductance and magnetic susceptibility measurements, electronic, FT-IR, EPR and (1)H NMR spectral studies. The cephalexin 2-pyridinylhydrazone ligand HL behaves as a monoanionic tridentate NNO chelating agent. The biological applications of complexes have been studied on three bacteria strains (Escherichia coli, Acinetobacter baumannii and Enterococcus faecalis) by agar diffusion disc method.
Collapse
Affiliation(s)
- J R Anacona
- Department of Chemistry, Universidad de Oriente, Cumana 6101 Venezuela.
| | - Maria Rincones
- Department of Chemistry, Universidad de Oriente, Cumana 6101 Venezuela
| |
Collapse
|
29
|
Schmuch J, Beckert S, Brandt S, Löhr G, Hermann F, Schmidt TJ, Beikler T, Hensel A. Extract from Rumex acetosa L. for prophylaxis of periodontitis: inhibition of bacterial in vitro adhesion and of gingipains of Porphyromonas gingivalis by epicatechin-3-O-(4β→8)-epicatechin-3-O-gallate (procyanidin-B2-Di-gallate). PLoS One 2015; 10:e0120130. [PMID: 25803708 PMCID: PMC4372542 DOI: 10.1371/journal.pone.0120130] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 01/19/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The aerial parts of Rumex acetosa L. have been used in traditional European medicine for inflammatory diseases of the mouth epithelial tissue. The following study aimed to investigate the influence of a proanthocyanidin-enriched extract from R. acetosa extract against the adhesion of Porphyromonas gingivalis (P. gingivalis), a pathogen strongly involved in chronic and aggressive periodontitis. A further goal was to define the bioactive lead structures responsible for a potential antiadhesive activity and to characterize the underlying molecular mechanisms of the antiadhesive effects. METHODOLOGY An extract of R. acetosa (RA1) with a defined mixture of flavan-3-ols, oligomeric proanthocyanidins and flavonoids, was used. Its impact on P. gingivalis adhesion to KB cells was studied by flow cytometry, confocal laser scanning microscopy and in situ adhesion assay using murine buccal tissue. RA1 and its compounds 1 to 15 were further investigated for additional effects on gingipain activity, hemagglutination and gene expression by RT-PCR. PRINCIPAL FINDINGS RA1 (5 to 15 μg/mL) reduced P. gingivalis adhesion in a dose-dependent manner to about 90%. Galloylated proanthocyanidins were confirmed to be responsible for this antiadhesive effect with epicatechin-3-O-gallate-(4β,8)-epicatechin-3'-O-gallate (syn. procyanidin B2-di-gallate) being the lead compound. Ungalloylated flavan-3-ols and oligomeric proanthocyanidins were inactive. RA1 and the galloylated proanthocyanidins strongly interact with the bacterial virulence factor Arg-gingipain, while the corresponding Lys-gingipain was hardly influenced. RA1 inhibited also hemagglutination. In silico docking studies indicated that epicatechin-3-O-gallate-(4β,8)-epicatechin-3'-O-gallate interacts with the active side of Arg-gingipain and hemaglutinin from P. gingivalis; the galloylation of the molecule seems to be responsible for fixation of the ligand to the protein. In conclusion, the proanthocyanidin-enriched extract RA1 and its main active constituent procyanidin B2-di-gallate protect cells from P. gingivalis infection by inhibiting bacterial adhesion to the host cell. RA1 and procyanidin B2-di-gallate appear to be promising candidates for future cytoprotective preparations for oral mouth care products.
Collapse
Affiliation(s)
- Jana Schmuch
- University of Münster, Institute for Pharmaceutical Biology and Phytochemistry, Münster, Germany
| | - Sabine Beckert
- University of Münster, Institute for Pharmaceutical Biology and Phytochemistry, Münster, Germany
| | - Simone Brandt
- University of Münster, Institute for Pharmaceutical Biology and Phytochemistry, Münster, Germany
| | - Gesine Löhr
- University of Münster, Institute for Pharmaceutical Biology and Phytochemistry, Münster, Germany
| | - Fabian Hermann
- University of Münster, Institute for Pharmaceutical Biology and Phytochemistry, Münster, Germany
| | - Thomas J. Schmidt
- University of Münster, Institute for Pharmaceutical Biology and Phytochemistry, Münster, Germany
| | - Thomas Beikler
- Heinrich-Heine-University, Department of Operative Dentistry, Periodontics and Endodontics, Düsseldorf, Germany
| | - Andreas Hensel
- University of Münster, Institute for Pharmaceutical Biology and Phytochemistry, Münster, Germany
| |
Collapse
|
30
|
Anacona JR, Noriega N, Camus J. Synthesis, characterization and antibacterial activity of a tridentate Schiff base derived from cephalothin and sulfadiazine, and its transition metal complexes. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2015; 137:16-22. [PMID: 25194315 DOI: 10.1016/j.saa.2014.07.091] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/06/2014] [Accepted: 07/29/2014] [Indexed: 06/03/2023]
Abstract
Metal(II) coordination compounds of a cephalothin Schiff base (H2L) derived from the condensation of cephalothin antibiotic with sulfadiazine were synthesized. The Schiff base ligand, mononuclear [ML(H2O)3] (M(II)=Mn,Co,Ni,Zn) complexes and magnetically diluted dinuclear copper(II) complex [CuL(H2O)3]2 were characterized by several techniques, including elemental and thermal analysis, molar conductance and magnetic susceptibility measurements, electronic, FT-IR, EPR and (1)H NMR spectral studies. The cephalothin Schiff base ligand H2L behaves as a dianionic tridentate NOO chelating agent. The biological applications of complexes have been studied on two bacteria strains (Escherichia coli and Staphylococcus aureus) by agar diffusion disc method.
Collapse
Affiliation(s)
- J R Anacona
- Department of Chemistry, Universidad de Oriente, Cumana 6101, Venezuela.
| | - Natiana Noriega
- Department of Chemistry, Universidad de Oriente, Cumana 6101, Venezuela
| | - Juan Camus
- Facultad de Ciencias Naturales y Exactas, Universidad de Playa Ancha, Valparaíso, Chile
| |
Collapse
|
31
|
Jarocki VM, Tacchi JL, Djordjevic SP. Non-proteolytic functions of microbial proteases increase pathological complexity. Proteomics 2015; 15:1075-88. [PMID: 25492846 PMCID: PMC7167786 DOI: 10.1002/pmic.201400386] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/26/2014] [Accepted: 12/05/2014] [Indexed: 12/26/2022]
Abstract
Proteases are enzymes that catalyse hydrolysis of peptide bonds thereby controlling the shape, size, function, composition, turnover and degradation of other proteins. In microbes, proteases are often identified as important virulence factors and as such have been targets for novel drug design. It is emerging that some proteases possess additional non‐proteolytic functions that play important roles in host epithelia adhesion, tissue invasion and in modulating immune responses. These additional “moonlighting” functions have the potential to obfuscate data interpretation and have implications for therapeutic design. Moonlighting enzymes comprise a subcategory of multifunctional proteins that possess at least two distinct biological functions on a single polypeptide chain. Presently, identifying moonlighting proteins relies heavily on serendipitous empirical data with clues arising from proteins lacking signal peptides that are localised to the cell surface. Here, we describe examples of microbial proteases with additional non‐proteolytic functions, including streptococcal pyrogenic exotoxin B, PepO and C5a peptidases, mycoplasmal aminopeptidases, mycobacterial chaperones and viral papain‐like proteases. We explore how these non‐proteolytic functions contribute to host cell adhesion, modulate the coagulation pathway, assist in non‐covalent folding of proteins, participate in cell signalling, and increase substrate repertoire. We conclude by describing how proteomics has aided in moonlighting protein discovery, focusing attention on potential moonlighters in microbial exoproteomes.
Collapse
Affiliation(s)
- Veronica M. Jarocki
- The ithree instituteProteomics Core Facility, University of TechnologySydneyNSWAustralia
| | - Jessica L. Tacchi
- The ithree instituteProteomics Core Facility, University of TechnologySydneyNSWAustralia
| | - Steven P. Djordjevic
- The ithree instituteProteomics Core Facility, University of TechnologySydneyNSWAustralia
- Proteomics Core FacilityUniversity of TechnologySydneyNSWAustralia
| |
Collapse
|
32
|
Jiang J, Liu HL, Liu ZH, Tan SW, Wu B. Identification of cystatin SN as a novel biomarker for pancreatic cancer. Tumour Biol 2015; 36:3903-10. [DOI: 10.1007/s13277-014-3033-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/30/2014] [Indexed: 12/14/2022] Open
|
33
|
Tamanai-Shacoori Z, Chandad F, Rébillard A, Cillard J, Bonnaure-Mallet M. Silver-zeolite combined to polyphenol-rich extracts of Ascophyllum nodosum: potential active role in prevention of periodontal diseases. PLoS One 2014; 9:e105475. [PMID: 25272151 PMCID: PMC4182675 DOI: 10.1371/journal.pone.0105475] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 07/21/2014] [Indexed: 01/18/2023] Open
Abstract
The purpose of this study was to evaluate various biological effects of silver-zeolite and a polyphenol-rich extract of A. nodosum (ASCOP) to prevent and/or treat biofilm-related oral diseases. Porphyromonas gingivalis and Streptococcus gordonii contribute to the biofilm formation associated with chronic periodontitis. In this study, we evaluated in vitro antibacterial and anti-biofilm effects of silver-zeolite (Ag-zeolite) combined to ASCOP on P. gingivalis and S. gordonii growth and biofilm formation capacity. We also studied the anti-inflammatory and antioxidant capacities of ASCOP in cell culture models. While Ag-zeolite combined with ASCOP was ineffective against the growth of S. gordonii, it showed a strong bactericidal effect on P. gingivalis growth. Ag-zeolite combined with ASCOP was able to completely inhibit S. gordonii monospecies biofilm formation as well as to reduce the formation of a bi-species S. gordonii/P. gingivalis biofilm. ASCOP alone was ineffective towards the growth and/or biofilm formation of S. gordonii and P. gingivalis while it significantly reduced the secretion of inflammatory cytokines (TNFα and IL-6) by LPS-stimulated human like-macrophages. It also exhibited antioxidant properties and decreased LPS induced lipid peroxidation in gingival epithelial cells. These findings support promising use of these products in future preventive or therapeutic strategies against periodontal diseases.
Collapse
Affiliation(s)
| | - Fatiha Chandad
- Groupe de Recherche en Ecologie Buccale, Faculté de médecine dentaire, Université Laval, Québec City, Québec, Canada
| | - Amélie Rébillard
- Laboratoire Mouvement, Sport, Santé, EA 1274, Université Rennes 1, Université Rennes 2, UEB, Rennes, France
| | - Josiane Cillard
- Laboratoire Mouvement, Sport, Santé, EA 1274, Université Rennes 1, Université Rennes 2, UEB, Rennes, France
| | - Martine Bonnaure-Mallet
- Equipe de Microbiologie, EA 1254, Université Rennes 1, UEB, Rennes, France
- Centre hospitalo-universitaire, Rennes, France
| |
Collapse
|
34
|
Olsen I, Potempa J. Strategies for the inhibition of gingipains for the potential treatment of periodontitis and associated systemic diseases. J Oral Microbiol 2014; 6:24800. [PMID: 25206939 PMCID: PMC4138498 DOI: 10.3402/jom.v6.24800] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/15/2014] [Accepted: 07/15/2014] [Indexed: 12/13/2022] Open
Abstract
Gingipains are the major virulence factors of Porphyromonas gingivalis, the main periodontopathogen. It is expected that inhibition of gingipain activity in vivo could prevent or slow down the progression of adult periodontitis. To date, several classes of gingipain inhibitors have been recognized. These include gingipain N-terminal prodomains, synthetic compounds, inhibitors from natural sources, antibiotics, antiseptics, antibodies, and bacteria. Several synthetic compounds are potent gingipain inhibitors but inhibit a broad spectrum of host proteases and have undesirable side effects. Synthetic compounds with high specificity for gingipains have unknown toxicity effects, making natural inhibitors more promising as therapeutic gingipain blockers. Cranberry and rice extracts interfere with gingipain activity and prevent the growth and biofilm formation of periodontopathogens. Although the ideal gingipain inhibitor has yet to be discovered, gingipain inhibition represents a novel approach to treat and prevent periodontitis. Gingipain inhibitors may also help treat systemic disorders that are associated with periodontitis, including cardiovascular disease, rheumatoid arthritis, aspiration pneumonia, pre-term birth, and low birth weight.
Collapse
Affiliation(s)
- Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland ; Department of Oral Immunology and Infectious Disease, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
35
|
Méndez Y, Pérez-Labrada K, González-Bacerio J, Valdés G, de los Chávez MÁ, Osuna J, Charli JL, Pascual I, Rivera DG. Combinatorial multicomponent access to natural-products-inspired peptidomimetics: discovery of selective inhibitors of microbial metallo-aminopeptidases. ChemMedChem 2014; 9:2351-9. [PMID: 24989844 DOI: 10.1002/cmdc.201402140] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Indexed: 11/09/2022]
Abstract
The development of selective inhibitors of microbial metallo-aminopeptidases is an important goal in the pursuit of antimicrobials for therapeutic applications. Herein, we disclose a combinatorial approach relying on two Ugi reactions for the generation of peptidomimetics inspired by natural metallo-aminopeptidase inhibitors. The library was screened for inhibitory activity against the neutral metallo-aminopeptidase of Escherichia coli (ePepN) and the porcine kidney cortex metallo-aminopeptidase (pAPN), which was used as a model of the M1-aminopeptidases of mammals. Six compounds showed typical dose-response inhibition profiles toward recombinant ePepN, with two of them being very potent and highly selective for ePepN over pAPN. Another compound showed moderate ePepN inhibition but total selectivity for this bacterial enzyme over its mammalian orthologue at concentrations of physiological relevance. This strategy proved to be useful for the identification of lead compounds for further optimization and development.
Collapse
Affiliation(s)
- Yanira Méndez
- Center for Natural Products Research, Faculty of Chemistry, University of Havana, Zapata y G, 10400, La Habana (Cuba)
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Dang HT, Chorell E, Uvell H, Pinkner JS, Hultgren SJ, Almqvist F. Syntheses and biological evaluation of 2-amino-3-acyl-tetrahydrobenzothiophene derivatives; antibacterial agents with antivirulence activity. Org Biomol Chem 2014; 12:1942-56. [PMID: 24531242 DOI: 10.1039/c3ob42478b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Developing new compounds targeting virulence factors (e.g., inhibition of pilus assembly by pilicides) is a promising approach to combating bacterial infection. A high-throughput screening campaign of a library of 17 500 small molecules identified 2-amino-3-acyl-tetrahydrobenzothiophene derivatives (hits 2 and 3) as novel inhibitors of pili-dependent biofilm formation in a uropathogenic Escherichia coli strain UTI89. Based on compounds 2 and 3 as the starting point, we designed and synthesized a series of structurally related analogs and investigated their activity against biofilm formation of E. coli UTI89. Systematic structural modification of the initial hits provided valuable information on their SARs for further optimization. In addition, small structural changes to the parent molecules resulted in low micromolar inhibitors (20-23) of E. coli biofilm development without an effect on bacterial growth. The hit compound 3 and its analog 20 were confirmed to prevent pili formation in a hemagglutination (HA) titer assay and electron microscopy (EM) measurements. These findings suggest that 2-amino-3-acyl-tetrahydrobenzothiophenes may serve as a new class of compounds for further elaboration as antibacterial agents with antivirulence activity.
Collapse
Affiliation(s)
- Hung The Dang
- Department of Chemistry, Umeå University, SE-90187 Umeå, Sweden.
| | | | | | | | | | | |
Collapse
|
37
|
Kim JT, Lee SJ, Kang MA, Park JE, Kim BY, Yoon DY, Yang Y, Lee CH, Yeom YI, Choe YK, Lee HG. Cystatin SN neutralizes the inhibitory effect of cystatin C on cathepsin B activity. Cell Death Dis 2013; 4:e974. [PMID: 24357805 PMCID: PMC3877556 DOI: 10.1038/cddis.2013.485] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 11/03/2013] [Accepted: 11/04/2013] [Indexed: 01/05/2023]
Abstract
Cystatin SN (CST1) is one of the several salivary cystatins that form tight equimolar complexes with cysteine proteases, such as the cathepsins. High expression of CST1 is correlated with advanced pTNM stage in gastric cancer. However, the functional role of CST1 in tumorigenesis has not been elucidated. In this study, we showed that CST1 was highly expressed in colon tumor tissues, compared with nontumor regions. Increased cell proliferation and invasiveness were observed in HCT116 cell lines stably transfected with CST1 cDNA (HCT116-CST1) but not in CST3-transfected cells. We also demonstrated that CST1-overexpressing cell lines exhibited increased tumor growth as well as metastasis in a xenograft nude mouse model. Interestingly, CST1 interacted with cystatin C (CST3), a potent cathepsin B (CTSB) inhibitor, with a higher affinity than the interaction between CST3 and CTSB in the extracellular space of HCT116 cells. CTSB-mediated cellular invasiveness and proteolytic activities were strongly inhibited by CST3, but in the presence of CST1 CTSB activities recovered significantly. Furthermore, domain mapping of CST1 showed that the disulfide-bonded conformation, or conserved folding, of CST1 is important for its secretion and for the neutralization of CST3 activity. These results suggest that CST1 upregulation might be involved in colorectal tumorigenesis and acts by neutralizing the inhibition of CTSB proteolytic activity by CST3.
Collapse
Affiliation(s)
- J-T Kim
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - S-J Lee
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - M A Kang
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - J E Park
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - B-Y Kim
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - D-Y Yoon
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Y Yang
- Department of Life Science, Sookmyung Women's University, Seoul, Republic of Korea
| | - C-H Lee
- Laboratory Animal Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Y I Yeom
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Y-K Choe
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - H G Lee
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| |
Collapse
|
38
|
Luminal cysteine-proteases degrade colonic tight junction structure and are responsible for abdominal pain in constipation-predominant IBS. Am J Gastroenterol 2013; 108:1322-31. [PMID: 23711626 DOI: 10.1038/ajg.2013.152] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/22/2013] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Luminal serine-proteases lead to increased colonic paracellular permeability and visceral hypersensitivity in patients with diarrhea-predominant irritable bowel syndrome (IBS-D). Other proteases, namely cysteine-proteases (CPs), increase airway permeability by digesting epithelial tight junction proteins. In this study, we focused on constipation-predominant IBS (IBS-C) and we aimed to (i) evaluate CP levels in two cohorts of IBS patients, (ii) test if IBS-C fecal supernatant (FSN) affects permeability, and visceral sensitivity after repeated administrations in mice, and (iii) evaluate occludin expression in IBS-C colonic biopsies. METHODS Fecal CP activity was determined using selective substrate and inhibitor (E64). The effect of papain, as positive control, and IBS-C FSN administrations were evaluated on colonic paracellular permeability and mucosal occludin levels in mice and T84 monolayers. Occludin protein levels were evaluated in IBS-C colonic biopsies. Sensitivity to colorectal distension (CRD) was measured after repeated administrations of IBS-C FSN. RESULTS We found in a subset of IBS-C patients an enhanced fecal CP activity, in comparison with healthy controls and IBS-D patients. CP activity levels positively correlated with disease severity and abdominal pain scoring. This association was confirmed by receiver operating characteristic curve analysis. In mice, repeated application of IBS-C FSN into colon triggered increased permeability, linked to the enzymatic degradation of occludin, and was associated with enhanced visceral sensitivity to CRD. Finally, occludin levels were found decreased in colonic biopsies from IBS-C patients, and IBS-C FSNs were able to degrade recombinant human occludin in vitro. All these effects were abolished by preincubation of IBS-C FSN with a CP inhibitor, E64. CONCLUSIONS These data suggest that luminal CPs may represent a new factor contributing to the genesis of symptoms in IBS.
Collapse
|
39
|
Fullagar JL, Garner AL, Struss AK, Day JA, Martin DP, Yu J, Cai X, Janda KD, Cohen SM. Antagonism of a zinc metalloprotease using a unique metal-chelating scaffold: tropolones as inhibitors of P. aeruginosa elastase. Chem Commun (Camb) 2013; 49:3197-9. [PMID: 23482955 PMCID: PMC3618488 DOI: 10.1039/c3cc41191e] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tropolone emerged from the screening of a chelator fragment library (CFL) as an inhibitor of the Zn(2+)-dependent virulence factor, Pseudomonas aeruginosa elastase (LasB). Based on this initial hit, a series of substituted tropolone-based LasB inhibitors was prepared, and a compound displaying potent activity in vitro and in a bacterial swarming assay was identified. Importantly, this inhibitor was found to be specific for LasB over other metalloenzymes, validating the usage of tropolone as a viable scaffold for identifying first-in-class LasB inhibitors.
Collapse
Affiliation(s)
- Jessica L. Fullagar
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA;Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA 92093
| | - Amanda L. Garner
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA;Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA 92093
| | - Anjali K. Struss
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA;Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA 92093
| | - Joshua A. Day
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA;Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA 92093
| | - David P. Martin
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA;Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA 92093
| | - Jing Yu
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA;Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA 92093
| | - Xiaoqing Cai
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA;Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA 92093
| | - Kim D. Janda
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA;Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA 92093
| | - Seth M. Cohen
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA;Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA 92093
| |
Collapse
|
40
|
Reactivity of mercury(II) halides with the α-keto stabilized sulfonium ylides: Crystal structures of two new polymer and binuclear complexes and in vitro antibacterial study. Polyhedron 2013. [DOI: 10.1016/j.poly.2012.10.054] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
41
|
Moitinho-Silva L, Kondo MY, Oliveira LCG, Okamoto DN, Paes JA, Machado MFM, Veronez CL, Motta G, Andrade SS, Juliano MA, Ferreira HB, Juliano L, Gouvea IE. Mycoplasma hyopneumoniae in vitro peptidase activities: identification and cleavage of kallikrein-kinin system-like substrates. Vet Microbiol 2013; 163:264-73. [PMID: 23421966 DOI: 10.1016/j.vetmic.2013.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 01/15/2013] [Accepted: 01/17/2013] [Indexed: 01/24/2023]
Abstract
Bacterial proteases are important for metabolic processes and pathogenesis in host organisms. The bacterial swine pathogen Mycoplasma hyopneumoniae has 15 putative protease-encoding genes annotated, but none of them have been functionally characterized. To identify and characterize peptidases that could be relevant for infection of swine hosts, we investigated the peptidase activity present in the pathogenic 7448 strain of M. hyopneumoniae. Combinatorial libraries of fluorescence resonance energy transfer peptides, specific inhibitors and pH profiling were used to screen and characterize endopeptidase, aminopeptidase and carboxypeptidase activities in cell lysates. One metalloendopeptidase, one serine endopeptidase, and one aminopeptidase were detected. The detected metalloendopeptidase activity, prominent at neutral and basic pH ranges, was due to a thimet oligopeptidase family member (M3 family), likely an oligoendopeptidase F (PepF), which cleaved the peptide Abz-GFSPFRQ-EDDnp at the F-S bond. A chymotrypsin-like serine endopeptidase activity, possibly a subtilisin-like serine protease, was prominent at higher pH levels, and was characterized by its preference for a Phe residue at the P1 position of the substrate. The aminopeptidase P (APP) activity showed a similar profile to that of human membrane-bound APP. Genes coding for these three peptidases were identified and their transcription was confirmed in the 7448 strain. Furthermore, M. hyopneumoniae cell lysate peptidases showed effects on kallikrein-kinin system-like substrates, such as bradykinin-derived substrates and human high molecular weight kininogen. The M. hyopneumoniae peptidase activities, here characterized for the first time, may be important for bacterial survival strategies and thus represent possible targets for drug development against M. hyopneumoniae swine infections.
Collapse
Affiliation(s)
- Lucas Moitinho-Silva
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Fröhlich E, Kantyka T, Plaza K, Schmidt KH, Pfister W, Potempa J, Eick S. Benzamidine derivatives inhibit the virulence of Porphyromonas gingivalis. Mol Oral Microbiol 2012; 28:192-203. [PMID: 23279840 DOI: 10.1111/omi.12015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2012] [Indexed: 12/12/2022]
Abstract
We have previously shown that benzamidine-type compounds can inhibit the activity of arginine-specific cysteine proteinases (gingipains HRgpA and RgpB); well-known virulence factors of Porphyromonas gingivalis. They also hinder in vitro growth of this important periodontopathogenic bacterium. Apparently growth arrest is not associated with their ability to inhibit these proteases, because pentamidine, which is a 20-fold less efficient inhibitor of gingipain than 2,6-bis-(4-amidinobenzyl)-cyclohexanone (ACH), blocked P. gingivalis growth far more effectively. To identify targets for benzamidine-derived compounds other than Arg-gingipains, and to explain their bacteriostatic effects, P. gingivalis ATCC 33277 and P. gingivalis M5-1-2 (clinical isolate) cell extracts were subjected to affinity chromatography using a benzamidine-Sepharose column to identify proteins interacting with benzamidine. In addition to HRgpA and RgpB the analysis revealed heat-shock protein GroEL as another ligand for benzamidine. To better understand the effect of benzamidine-derived compounds on P. gingivalis, bacteria were exposed to benzamidine, pentamidine, ACH and heat, and the expression of gingipains and GroEL was determined. Exposure to heat and benzamidine-derived compounds caused significant increases in GroEL, at both the mRNA and protein levels. Interestingly, despite the fact that gingipains were shown to be the main virulence factors in a fertilized egg model of infection, mortality rates were strongly reduced, not only by ACH, but also by pentamidine, a relatively weak gingipain inhibitor. This effect may depend not only on gingipain inhibition but also on interaction of benzamidine derivatives with GroEL. Therefore these compounds may find use in supportive periodontitis treatment.
Collapse
Affiliation(s)
- E Fröhlich
- Department of Experimental Anesthesiology, Clinic for Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Alginate lyase exhibits catalysis-independent biofilm dispersion and antibiotic synergy. Antimicrob Agents Chemother 2012; 57:137-45. [PMID: 23070175 DOI: 10.1128/aac.01789-12] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
More than 2 decades of study support the hypothesis that alginate lyases are promising therapeutic candidates for treating mucoid Pseudomonas aeruginosa infections. In particular, the enzymes' ability to degrade alginate, a key component of mucoid biofilm matrix, has been the presumed mechanism by which they disrupt biofilms and enhance antibiotic efficacy. The systematic studies reported here show that, in an in vitro model, alginate lyase dispersion of P. aeruginosa biofilms and enzyme synergy with tobramycin are completely decoupled from catalytic activity. In fact, equivalent antibiofilm effects can be achieved with bovine serum albumin or simple amino acids. These results provide new insights into potential mechanisms of alginate lyase therapeutic activity, and they should motivate a careful reexamination of the fundamental assumptions underlying interest in enzymatic biofilm dispersion.
Collapse
|
44
|
Garner AL, Struss AK, Fullagar JL, Agrawal A, Moreno AY, Cohen SM, Janda KD. 3-Hydroxy-1-alkyl-2-methylpyridine-4(1H)-thiones: Inhibition of the Pseudomonas aeruginosa Virulence Factor LasB. ACS Med Chem Lett 2012. [PMID: 23181168 DOI: 10.1021/ml300128f] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Bacterial resistance coupled to our current arsenal of antibiotics presents us with a growing threat to public health, thus warranting the exploration of alternative antibacterial strategies. In particular, the targeting of virulence factors has been regarded as a "second generation" antibiotic approach. In Pseudomonas aeruginosa, a Zn(2+) metalloprotease virulence factor, LasB or P. aeruginosa elastase, has been implicated in the development of P. aeruginosa-related keratitis, pneumonia and burn infection. Moreover, the enzyme also plays a critical role in swarming and biofilm formation, both of which are processes that have been linked to antibiotic resistance. To further validate the importance of LasB in P. aeruginosa infection, we describe our efforts toward the discovery of non-peptidic small molecule inhibitors of LasB. Using identified compounds, we have confirmed the role that LasB plays in P. aeruginosa swarming and demonstrate the potential for LasB-targeted small molecules in studying antimicrobial resistant P. aeruginosa phenotypes.
Collapse
Affiliation(s)
- Amanda L. Garner
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| | - Anjali K. Struss
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| | - Jessica L. Fullagar
- Department of Chemistry and
Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, United States
| | - Arpita Agrawal
- Department of Chemistry and
Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, United States
| | - Amira Y. Moreno
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| | - Seth M. Cohen
- Department of Chemistry and
Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, United States
| | - Kim D. Janda
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| |
Collapse
|
45
|
|
46
|
Specific interactions and binding energies between thermolysin and potent inhibitors: Molecular simulations based on ab initio molecular orbital method. J Mol Graph Model 2012; 33:1-11. [DOI: 10.1016/j.jmgm.2011.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 10/18/2011] [Accepted: 10/18/2011] [Indexed: 11/19/2022]
|
47
|
Wildeboer D, Hill KE, Jeganathan F, Williams DW, Riddell AD, Price PE, Thomas DW, Stephens P, Abuknesha RA, Price RG. Specific protease activity indicates the degree of Pseudomonas aeruginosa infection in chronic infected wounds. Eur J Clin Microbiol Infect Dis 2012; 31:2183-9. [DOI: 10.1007/s10096-012-1553-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 01/07/2012] [Indexed: 11/24/2022]
|
48
|
Microbial and fungal protease inhibitors--current and potential applications. Appl Microbiol Biotechnol 2012; 93:1351-75. [PMID: 22218770 PMCID: PMC7080157 DOI: 10.1007/s00253-011-3834-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Revised: 12/07/2011] [Accepted: 12/09/2011] [Indexed: 01/18/2023]
Abstract
Proteolytic enzymes play essential metabolic and regulatory functions in many biological processes and also offer a wide range of biotechnological applications. Because of their essential roles, their proteolytic activity needs to be tightly regulated. Therefore, small molecules and proteins that inhibit proteases can be versatile tools in the fields of medicine, agriculture and biotechnology. In medicine, protease inhibitors can be used as diagnostic or therapeutic agents for viral, bacterial, fungal and parasitic diseases as well as for treating cancer and immunological, neurodegenerative and cardiovascular diseases. They can be involved in crop protection against plant pathogens and herbivorous pests as well as against abiotic stress such as drought. Furthermore, protease inhibitors are indispensable in protein purification procedures to prevent undesired proteolysis during heterologous expression or protein extraction. They are also valuable tools for simple and effective purification of proteases, using affinity chromatography. Because there are such a large number and diversity of proteases in prokaryotes, yeasts, filamentous fungi and mushrooms, we can expect them to be a rich source of protease inhibitors as well.
Collapse
|
49
|
Vincents B, Guentsch A, Kostolowska D, von Pawel-Rammingen U, Eick S, Potempa J, Abrahamson M. Cleavage of IgG1 and IgG3 by gingipain K from Porphyromonas gingivalis may compromise host defense in progressive periodontitis. FASEB J 2011; 25:3741-50. [PMID: 21768393 PMCID: PMC3177567 DOI: 10.1096/fj.11-187799] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 07/01/2011] [Indexed: 01/08/2023]
Abstract
Degradation of immunoglobulins is an effective strategy of bacteria to evade the immune system. We have tested whether human IgG is a substrate for gingipain K of Porphyromonas gingivalis and found that the enzyme can hydrolyze subclass 1 and 3 of human IgG. The heavy chain of IgG(1) was cleaved at a single site within the hinge region, generating Fab and Fc fragments. IgG(3) was also cleaved within the heavy chain, but at several sites around the CH2 region. Investigation of the enzyme kinetics of IgG proteolysis by gingipain K, using FPLC- and isothermal titration calorimetry-based assays followed by Hill plots, revealed non-Michaelis-Menten kinetics involving a mechanism of positive cooperativity. In ex vivo studies, it was shown that gingipain K retained its IgG hydrolyzing activity in human plasma despite the high content of natural protease inhibitors; that IgG(1) cleavage products were detected in gingival crevicular fluid samples from patients with severe periodontitis; and that gingipain K treatment of serum samples from patients with high antibody titers against P. gingivalis significantly hindered opsonin-dependent phagocytosis of clinical isolates of P. gingivalis by neutrophils. Altogether, these findings underline a biological function of gingipain K as an IgG protease of pathophysiological importance.
Collapse
Affiliation(s)
- Bjarne Vincents
- Department of Laboratory Medicine, Division of Clinical Chemistry and Pharmacology, Lund University, University Hospital, Lund, Sweden
| | - Arndt Guentsch
- Department of Conservative Dentistry, University Hospital of Jena, Jena, Germany
| | - Dominika Kostolowska
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Kraków, Poland
| | | | - Sigrun Eick
- Department of Periodontology, Laboratory of Oral Microbiology, University of Bern, Bern, Switzerland; and
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Kraków, Poland
- University of Louisville Dental School, Center for Oral Health and Systemic Diseases, Louisville, Kentucky, USA
| | - Magnus Abrahamson
- Department of Laboratory Medicine, Division of Clinical Chemistry and Pharmacology, Lund University, University Hospital, Lund, Sweden
| |
Collapse
|
50
|
Al-Amiery AA, Al-Bayati RIH, Saour KY, Radi MF. Cytotoxicity, antioxidant, and antimicrobial activities of novel 2-quinolone derivatives derived from coumarin. RESEARCH ON CHEMICAL INTERMEDIATES 2011. [DOI: 10.1007/s11164-011-0371-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|