1
|
Dewan D, Basu A, Dolai D, Pal S. Biological and Biophysical Methods for Evaluation of Inhibitors of Sortase A in Staphylococcus aureus: An Overview. Cell Biochem Funct 2024; 42:e70002. [PMID: 39470102 DOI: 10.1002/cbf.70002] [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: 06/15/2024] [Revised: 09/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Staphylococcus aureus, one of the most notorious pathogens, develops antibiotic resistance by the formation of a thick layer of exopolysaccharides known as biofilms. Sortase A, a transpeptidase responsible for biofilm formation and attachment to the host surface, has emerged as an important drug target for development of anti-virulence agents. A number of sortase A inhibitors, both peptide and non-peptides are reported which involved the use of several experiments which may provide insights regarding binding affinity, specificity, safety, and efficacy of ligands. In this review, we focus on the principles, pros and cons, and the type of information obtained from biophysical (FRET assay, Microscale Thermophoresis, Surface Plasmon Resonance, CD spectroscopy etc.) and biological (cell viability assay, biofilm formation assay, CLSM, western blot analysis, in vivo characterization on mice etc.) methods for estimation of probable sortase A inhibitors, which might be helpful to the researchers who might be interested to delve into the development of sortase A inhibitors as a drug, to address the burning question of antimicrobial resistance (AMR).
Collapse
|
2
|
Benito-Vázquez I, Muñoz-Labrador A, Garrido-Romero M, Hontoria-Caballo G, García-García C, Diez-Municio M, Moreno FJ. New Pipeline for Analysing Fruit Proteolytic Products Used as Digestive Health Nutraceuticals. Int J Mol Sci 2024; 25:10315. [PMID: 39408644 PMCID: PMC11476805 DOI: 10.3390/ijms251910315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 10/20/2024] Open
Abstract
Proteolytic products are extensively used in the nutraceutical sector to improve protein digestion and muscle quality in target populations (e.g., athletes or elderly). These products are processed using techniques that often lead to low purity but competitive pricing. Despite their widespread use and well-established production methods, the industry lacks standardized analytical methods for assessing these products and detecting potential fraud. This study proposes a comprehensive and harmonized pipeline for their analysis, which includes quantifying total soluble protein and proteolytic activity, as well as the determination of product stability and protein profile using SDS-PAGE and proteomic techniques. Despite the fact that protease extracts from pineapple had the highest protein content, most of the bromelain remained inactive, unlike in kiwi and papaya. SDS-PAGE revealed partial protein degradation of pineapple extracts, whereas kiwi extracts reflected a lower purification level but a higher protein integrity. The application of proteomic approaches strengthened the identification and origin tracing of the proteases. This study contributes to the development of a robust framework for analyzing proteolytic extracts, spanning from soluble protein quantification to protein profiling and activity determination. It may also ensure reliable supplier selection, high-quality manufacturing practices, and the implementation of optimal storage and formulation strategies in the nutraceutical industry.
Collapse
Affiliation(s)
- Iván Benito-Vázquez
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain; (I.B.-V.); (A.M.-L.); (M.G.-R.)
- Pharmactive Biotech Products SLU, Faraday 7, 28049 Madrid, Spain;
| | - Ana Muñoz-Labrador
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain; (I.B.-V.); (A.M.-L.); (M.G.-R.)
| | - Manuel Garrido-Romero
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain; (I.B.-V.); (A.M.-L.); (M.G.-R.)
- Pharmactive Biotech Products SLU, Faraday 7, 28049 Madrid, Spain;
| | | | - Carlos García-García
- Centro de Biología Molecular Severo Ochoa, CBM (CSIC-UAM), Nicolás Cabrera, 1, 28049 Madrid, Spain;
| | | | - F. Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain; (I.B.-V.); (A.M.-L.); (M.G.-R.)
| |
Collapse
|
3
|
Diogo MA, Cabral AGT, de Oliveira RB. Advances in the Search for SARS-CoV-2 M pro and PL pro Inhibitors. Pathogens 2024; 13:825. [PMID: 39452697 PMCID: PMC11510351 DOI: 10.3390/pathogens13100825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/19/2024] [Accepted: 09/22/2024] [Indexed: 10/26/2024] Open
Abstract
SARS-CoV-2 is a spherical, positive-sense, single-stranded RNA virus with a large genome, responsible for encoding both structural proteins, vital for the viral particle's architecture, and non-structural proteins, critical for the virus's replication cycle. Among the non-structural proteins, two cysteine proteases emerge as promising molecular targets for the design of new antiviral compounds. The main protease (Mpro) is a homodimeric enzyme that plays a pivotal role in the formation of the viral replication-transcription complex, associated with the papain-like protease (PLpro), a cysteine protease that modulates host immune signaling by reversing post-translational modifications of ubiquitin and interferon-stimulated gene 15 (ISG15) in host cells. Due to the importance of these molecular targets for the design and development of novel anti-SARS-CoV-2 drugs, the purpose of this review is to address aspects related to the structure, mechanism of action and strategies for the design of inhibitors capable of targeting the Mpro and PLpro. Examples of covalent and non-covalent inhibitors that are currently being evaluated in preclinical and clinical studies or already approved for therapy will be also discussed to show the advances in medicinal chemistry in the search for new molecules to treat COVID-19.
Collapse
Affiliation(s)
| | | | - Renata Barbosa de Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (M.A.D.); (A.G.T.C.)
| |
Collapse
|
4
|
Malatyali E, Guclu O, Yildiz I, Ertug S, Ertabaklar H. The Low Genetic Diversity of Dientamoeba fragilis Isolates in Southwest Turkey and Analysis of Clinical Findings. Acta Parasitol 2024; 69:1576-1586. [PMID: 39164548 DOI: 10.1007/s11686-024-00896-x] [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: 02/18/2024] [Accepted: 07/30/2024] [Indexed: 08/22/2024]
Abstract
PURPOSE Dientamoeba fragilis (D. fragilis) is a common intestinal protozoan with a global distribution. In the present study, we aimed to determine genetic diversity of D. fragilis isolates with multilocus sequence typing (MLST) in the southwest of Turkey and analyse the clinical findings. MATERIALS AND METHODS The study included faecal samples from 200 individuals in Aydin, Turkey. The positivity of D. fragilis was determined with 18 S rRNA gene-based PCR assay. Six nested-PCR reactions were set to amplify partial D. fragilis housekeeping genes in the positive samples. The sequences were aligned with the references from GenBank to detect nucleotide polymorphisms and haplotypes. Additionally, the clinical findings and demographic characteristics of patients were statistically analysed between D. fragilis-infected and non-infected cases. RESULTS The positivity of D. fragilis was 16% (32 out of 200 cases) with 18 S rRNA based-PCR, and all were classified as "genotype 1". The analysis of six MLST loci revealed different haplotypes only at one locus; the remaining five loci exhibited no polymorphisms. The haplotypes in the present study were identical to at least one previously reported reference, except the locus "large subunit of RNA polymerase II" locus. There were no significant differences in any of the clinical findings or demographic characteristics between the infected and non-infected groups. CONCLUSIONS Our study revealed a low genetic diversity of D. fragilis isolates from Turkey, like other countries including Italy, Denmark, the UK, Australia, and Brazil. The high degree of sequence similarity in housekeeping genes indicated the clonal distribution of D. fragilis.
Collapse
Affiliation(s)
- Erdogan Malatyali
- Department of Parasitology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, 09100, Turkey.
| | - Ozgur Guclu
- Department of Plant and Animal Production, Aydin Adnan Menderes University, Sultanhisar MYO, Aydin, 09100, Turkey
- Recombinant DNA and Recombinant Protein Research Centre (REDPROM), Aydin Adnan Menderes University, Aydin, 09100, Turkey
| | - Ibrahim Yildiz
- Department of Parasitology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, 09100, Turkey
| | - Sema Ertug
- Department of Parasitology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, 09100, Turkey
| | - Hatice Ertabaklar
- Department of Parasitology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, 09100, Turkey
| |
Collapse
|
5
|
Li CC, Lin CC, Lee CY, Sheu ML, Tsai YC, Tsai CY, Wu HT, Wu RJ, Lai DW. Therapeutic Effect of Lecigel, Cetiol ®CC, Activonol-6, Activonol-M, 1,3-Propanediol, Soline, and Fucocert ® (LCAA-PSF) Treatment on Imiquimod-Induced Psoriasis-like Skin in Mice. Int J Mol Sci 2024; 25:7720. [PMID: 39062965 PMCID: PMC11276952 DOI: 10.3390/ijms25147720] [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: 06/06/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
The individual ingredients of 1,3-Propanediol, Soline, and Fucocert® (PSF) are often used as cosmetic formulations in skin care. In addition, the mixture of Lecigel, Cetiol®CC, Activonol-6, and Activonol-M (LCAA) is often used as a cosmetic base. However, whether the combination of LCAA with PSF (LCAA-PSF) exerts a therapeutic effect on psoriasis remains unclear. In this study, mice induced with imiquimod (IMQ) were divided into three groups and administered 100 mg/day of LCAA, 100 mg/day of LCAA-PSF, or Vaseline on the dorsal skin of each mouse. Weight-matched mice treated with Vaseline alone were used as controls. Hematoxylin and eosin (H&E) staining and enzyme-linked immunosorbent assay(ELISA) were used to assess tissue morphology and inflammatory cytokines. RNA sequencing analysis was used to predict the mechanism underlying the action of LCAA-PSF against psoriasis, while immunohistochemical analysis validation was used to identify pertinent molecular pathways. The results demonstrated that LCAA-PSF alleviated IMQ-induced keratinocyte differentiation/ proliferation bydecreasingthe serum levels of inflammatory cytokines such as IL-6, TNF-α, IL-23, and IL-17A and the epidermisof TGFβ, Ki67, CK5/6, and VEGF expression, which is associated with angiogenesis and keratinocyte differentiation/ proliferation. These findings highlight the antipsoriatic activity of LCAA-PSF in a psoriasis-like mouse model and suggest this may occurvia the inhibition of inflammatory factor secretionand the TGFβ-related signal pathway.
Collapse
Affiliation(s)
- Chih-Ching Li
- Department of Applied Chemistry, Providence University, 200, Sec. 7, Taiwan Boulevard, Shalu Dist., Taichung 43301, Taiwan;
- Department of Pediatrics, Chang Bing Show Chwan Memorial Hospital, No. 6, Lugong Rd. Lugang Township, Changhua 505029, Taiwan;
| | - Chih-Chien Lin
- Department of Cosmetic Science, Providence University, 200, Sec. 7, Taiwan Boulevard, Shalu Dist., Taichung 43301, Taiwan;
| | - Chun-Yi Lee
- Department of Pediatrics, Chang Bing Show Chwan Memorial Hospital, No. 6, Lugong Rd. Lugang Township, Changhua 505029, Taiwan;
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan;
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Yi-Ching Tsai
- Immunomedicine Group, Department of Molecular Biology and Cell Research, Chang Bing Show Chwan Memorial Hospital, Changhua 505029, Taiwan;
| | - Chia-Yun Tsai
- Experimental Animal Center, Department of Molecular Biology and Cell Research, Chang Bing Show Chwan Memorial Hospital, Changhua 505029, Taiwan; (C.-Y.T.); (H.-T.W.)
| | - Hao-Ting Wu
- Experimental Animal Center, Department of Molecular Biology and Cell Research, Chang Bing Show Chwan Memorial Hospital, Changhua 505029, Taiwan; (C.-Y.T.); (H.-T.W.)
| | - Ren-Jang Wu
- Department of Applied Chemistry, Providence University, 200, Sec. 7, Taiwan Boulevard, Shalu Dist., Taichung 43301, Taiwan;
| | - De-Wei Lai
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Experimental Animal Center, Department of Molecular Biology and Cell Research, Chang Bing Show Chwan Memorial Hospital, Changhua 505029, Taiwan; (C.-Y.T.); (H.-T.W.)
- Department of Pharmacy and Master Program, Tajen University, Pingtung 907101, Taiwan
| |
Collapse
|
6
|
Liatsou I, Assefa B, Liyanage W, Surasinghe S, Nováková Z, Bařinka C, Gabrielson K, Raman V, Artemov D, Hapuarachchige S. Development and therapeutic evaluation of 5D3(CC-MLN8237) 3.2 antibody-theranostic conjugates for PSMA-positive prostate cancer therapy. Front Pharmacol 2024; 15:1385598. [PMID: 38751786 PMCID: PMC11094276 DOI: 10.3389/fphar.2024.1385598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Prostate cancer (PC) is an aggressive cancer that can progress rapidly and eventually become castrate-resistant prostate cancer (CRPC). Stage IV metastatic castrate-resistant prostate cancer (mCRPC) is an incurable late-stage cancer type with a low 5-year overall survival rate. Targeted therapeutics such as antibody-drug conjugates (ADCs) based on high-affinity monoclonal antibodies and potent drugs conjugated via smart linkers are being developed for PC management. Conjugating further with in vitro or in vivo imaging agents, ADCs can be used as antibody-theranostic conjugates (ATCs) for diagnostic and image-guided drug delivery. In this study, we have developed a novel ATC for PSMA (+) PC therapy utilizing (a) anti-PSMA 5D3 mAb, (b) Aurora A kinase inhibitor, MLN8237, and (c) for the first time using tetrazine (Tz) and trans-cyclooctene (TCO) click chemistry-based conjugation linker (CC linker) in ADC development. The resulting 5D3(CC-MLN8237)3.2 was labeled with suitable fluorophores for in vitro and in vivo imaging. The products were characterized by SDS-PAGE, MALDI-TOF, and DLS and evaluated in vitro by optical imaging, flow cytometry, and WST-8 assay for cytotoxicity in PSMA (+/-) cells. Therapeutic efficacy was determined in human PC xenograft mouse models following a designed treatment schedule. After the treatment study animals were euthanized, and toxicological studies, complete blood count (CBC), blood clinical chemistry analysis, and H&E staining of vital organs were conducted to determine side effects and systemic toxicities. The IC50 values of 5D3(CC-MLN8237)3.2-AF488 in PSMA (+) PC3-PIP and PMSA (-) PC3-Flu cells are 8.17 nM and 161.9 nM, respectively. Pure MLN8237 shows 736.9 nM and 873.4 nM IC50 values for PC3-PIP and PC3-Flu cells, respectively. In vivo study in human xenograft mouse models confirmed high therapeutic efficacy of 5D3(CC-MLN8237)3.2-CF750 with significant control of PSMA (+) tumor growth with minimal systemic toxicity in the treated group compared to PSMA (-) treated and untreated groups. Approximately 70% of PSMA (+) PC3-PIP tumors did not exceed the threshold of the tumor size in the surrogate Kaplan-Meyer analysis. The novel ATC successfully controlled the growth of PSMA (+) tumors in preclinical settings with minimal systemic toxicities. The therapeutic efficacy and favorable safety profile of novel 5D3(CC-MLN8237)3.2 ATC demonstrates their potential use as a theranostic against aggressive PC.
Collapse
Affiliation(s)
- Ioanna Liatsou
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Betelhem Assefa
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wathsala Liyanage
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sharmane Surasinghe
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Zora Nováková
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Cyril Bařinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Venu Raman
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dmitri Artemov
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sudath Hapuarachchige
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
7
|
Wu H, Jmel MA, Chai J, Tian M, Xu X, Hui Y, Nandakumar KS, Kotsyfakis M. Tick cysteine protease inhibitors suppress immune responses in mannan-induced psoriasis-like inflammation. Front Immunol 2024; 15:1344878. [PMID: 38444844 PMCID: PMC10912570 DOI: 10.3389/fimmu.2024.1344878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Protease inhibitors regulate various biological processes and prevent host tissue/organ damage. Specific inhibition/regulation of proteases is clinically valuable for treating several diseases. Psoriasis affects the skin in the limbs and scalp of the body, and the contribution of cysteine and serine proteases to the development of skin inflammation is well documented. Cysteine protease inhibitors from ticks have high specificity, selectivity, and affinity to their target proteases and are efficient immunomodulators. However, their potential therapeutic effect on psoriasis pathogenesis remains to be determined. Therefore, we tested four tick cystatins (Sialostatin L, Sialostatin L2, Iristatin, and Mialostatin) in the recently developed, innate immunity-dependent mannan-induced psoriasis model. We explored the effects of protease inhibitors on clinical symptoms and histological features. In addition, the number and percentage of immune cells (dendritic cells, neutrophils, macrophages, and γδT cells) by flow cytometry, immunofluorescence/immunohistochemistry and, the expression of pro-inflammatory cytokines (TNF-a, IL-6, IL-22, IL-23, and IL-17 family) by qPCR were analyzed using skin, spleen, and lymph node samples. Tick protease inhibitors have significantly decreased psoriasis symptoms and disease manifestations but had differential effects on inflammatory responses and immune cell populations, suggesting different modes of action of these inhibitors on psoriasis-like inflammation. Thus, our study demonstrates, for the first time, the usefulness of tick-derived protease inhibitors for treating skin inflammation in patients.
Collapse
Affiliation(s)
- Huimei Wu
- Department of Pharmacy, The Eighth Affiliated City Hospital of Guangzhou Medical University, The Eighth People’s Hospital of Guangzhou, Guangzhou, China
- Karolinska Institute United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Mohamed Amine Jmel
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czechia
| | - Jinwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Maolin Tian
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Hui
- Department of Endocrinology, Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Kutty Selva Nandakumar
- Karolinska Institute United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Department of Environmental and Biosciences, School of Business, Innovation and Sustainability, Halmstad University, Halmstad, Sweden
| | - Michail Kotsyfakis
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czechia
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| |
Collapse
|