1
|
Bae HE, Jung J, Sung JS, Kwon S, Kang MJ, Jose J, Lee M, Pyun JC. Screening of deoxyribonuclease I inhibitors from autodisplayed Fv-antibody library. Int J Biol Macromol 2025; 304:140770. [PMID: 39922350 DOI: 10.1016/j.ijbiomac.2025.140770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Deoxyribonuclease (DNase) I inhibitors have been developed based on proteins, nucleotides and synthetic compounds. In this work, amino acid sequences with the activity of DNase I inhibitor were screened from an Fv-antibody library expressed on the outer membrane of Escherichia coli. The Fv-antibody indicated the heavy chain variable region (VH) of immunoglobulin G (IgG) and the Fv-antibody library was generated with a randomized complementarity-determining region 3 (CDR3). From the Fv-antibody library, two clones were screened for their binding affinity to DNase I and expressed as soluble recombinant proteins as well as peptides. The binding affinity (KD) to DNase I was estimated for the expressed Fv-antibodies (73.4 nM for Fv-1 and 89.0 nM for Fv-19) and synthesized peptides (279.2 nM for Peptide-1 and 243.2 nM for Peptide-19) using SPR biosensor. The inhibitory activity (IC50) of the expressed Fv-antibodies (550.0 nM for Fv-1 and 660.2 nM for Fv-19) and synthetic peptides (864.5 nM for Peptide-1 and 974.6 nM for Peptide-19) was measured using agarose-gel assay and TaqMan-like fluorescence assay. These IC50 values indicated that both expressed Fv-antibodies and synthesized peptides exerted an effective inhibitory activity against DNase I. The interaction between the screened inhibitors and DNase I was analyzed by docking simulation.
Collapse
Affiliation(s)
- Hyung Eun Bae
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Jaeyong Jung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Jeong Soo Sung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Soonil Kwon
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Joachim Jose
- Institute of Pharmaceutical and Medical Chemistry, Westfälischen Wilhelms-Universität Münster, Muenster 48149, Germany
| | - Misu Lee
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
2
|
Volek M, Kurfürst J, Kožíšek M, Srb P, Veverka V, Curtis E. Apollon: a deoxyribozyme that generates a yellow product. Nucleic Acids Res 2024; 52:9062-9075. [PMID: 38869058 PMCID: PMC11347176 DOI: 10.1093/nar/gkae490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024] Open
Abstract
Colorimetric assays in which the color of a solution changes in the presence of an input provide a simple and inexpensive way to monitor experimental readouts. In this study we used in vitro selection to identify a self-phosphorylating kinase deoxyribozyme that produces a colorimetric signal by converting the colorless substrate pNPP into the yellow product pNP. The minimized catalytic core, sequence requirements, secondary structure, and buffer requirements of this deoxyribozyme, which we named Apollon, were characterized using a variety of techniques including reselection experiments, high-throughput sequencing, comparative analysis, biochemical activity assays, and NMR. A bimolecular version of Apollon catalyzed multiple turnover phosphorylation and amplified the colorimetric signal. Engineered versions of Apollon could detect oligonucleotides with specific sequences as well as several different types of nucleases in homogenous assays that can be performed in a single tube without the need for washes or purifications. We anticipate that Apollon will be particularly useful to reduce costs in high-throughput screens and for applications in which specialized equipment is not available.
Collapse
Affiliation(s)
- Martin Volek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
- Department of Genetics and Microbiology, Faculty of Science, Charles University in Prague, Prague 128 44, Czech Republic
| | - Jaroslav Kurfürst
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
- Department of Informatics and Chemistry, University of Chemistry and Technology, Prague 166 28, Czech Republic
| | - Milan Kožíšek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| | - Pavel Srb
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| | - Václav Veverka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University in Prague, Prague 128 44, Czech Republic
| | - Edward A Curtis
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| |
Collapse
|
3
|
Morita K, Moriwaki T, Habe S, Taniguchi-Ikeda M, Hasegawa T, Minato Y, Aoi T, Maruyama T. Molecular Aggregation Strategy for Inhibiting DNases. JACS AU 2024; 4:2262-2266. [PMID: 38938790 PMCID: PMC11200219 DOI: 10.1021/jacsau.4c00210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 06/29/2024]
Abstract
This study highlights the novel potential of molecular aggregates as inhibitors of a disease-related protein. Enzyme inhibitors have been studied and developed as molecularly targeted drugs and have been applied for cancer, autoimmune diseases, and infections. In many cases, enzyme inhibitors that are used for therapeutic applications interact directly with enzymes in a molecule-to-molecule manner. We found that the aggregates of a small compound, Mn007, inhibited bovine pancreatic DNase I. Once Mn007 molecules formed aggregates, they exhibited inhibitory effects specific to DNases that require divalent metal ions. A DNase secreted by Streptococcus pyogenes causes streptococcal toxic shock syndrome (STSS). STSS is a severe infectious disease with a fatality rate exceeding 30% in patients, even in this century. S. pyogenes disrupts the human barrier system against microbial infections through the secreted DNase. Until now, the discovery/development of a DNase inhibitor has been challenging. Mn007 aggregates were found to inhibit the DNase secreted by S. pyogenes, which led to the successful suppression of S. pyogenes growth in human whole blood. To date, molecular aggregation has been outside the scope of drug discovery. The present study suggests that molecular aggregation is a vast area to be explored for drug discovery and development because aggregates of small-molecule compounds can inhibit disease-related enzymes.
Collapse
Affiliation(s)
- Kenta Morita
- Department
of Chemical Science and
Engineering, Graduate School of Engineering, Kobe University 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
- Research
Center for Membrane and Film Technology, Kobe University, 1-1
Rokkodai, Nada-ku, Kobe 657-8501, Japan
| | - Tomoko Moriwaki
- Department
of Chemical Science and
Engineering, Graduate School of Engineering, Kobe University 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
| | - Shunsuke Habe
- Department
of Chemical Science and
Engineering, Graduate School of Engineering, Kobe University 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
| | - Mariko Taniguchi-Ikeda
- Department
of Clinical Genetics, Fujita Health University
Hospital 1-98 Dengakugakubo, Kutsukake-chou, Toyoake, Aichi 470-1192, Japan
| | - Tadao Hasegawa
- Department
of Bacteriology, Graduate School of Medical
Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Yusuke Minato
- Department
of Microbiology, School of Medicine, Fujita
Health University, 1-98
Dengakugakubo, Kutsukake-chou, Toyoake, Aichi 470-1192, Japan
| | - Takashi Aoi
- Division
of Stem Cell Medicine, Graduate School of Medicine, Kobe University, 7-5-1
Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tatsuo Maruyama
- Department
of Chemical Science and
Engineering, Graduate School of Engineering, Kobe University 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
- Research
Center for Membrane and Film Technology, Kobe University, 1-1
Rokkodai, Nada-ku, Kobe 657-8501, Japan
| |
Collapse
|
4
|
Konwar B, De S, Das G, Ramesh A. Inhibition of staphylococcal nuclease by benzimidazole-based Ligand: Implications in DNA-Mediated entrapment and uptake of MRSA by Macrophage-like cells. Bioorg Chem 2024; 144:107133. [PMID: 38278047 DOI: 10.1016/j.bioorg.2024.107133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/22/2023] [Accepted: 01/11/2024] [Indexed: 01/28/2024]
Abstract
The staphylococcal nuclease also referred as micrococcal nuclease (MNase) is a key drug target as the enzyme degrades the neutrophil extracellular trap (NET) and empowers the pathogen to subvert the host innate immune system. To this end, the current study presents a critical evaluation of MNase inhibition rendered by benzimidazole-based ligands (C1 and C2) and probes its therapeutic implications. A nuclease assay indicated that MNase inhibition rendered by C1 and C2 was ∼ 55 % and ∼ 72 %, respectively, at the highest tested concentration of 10 µM. Studies on enzyme kinetics revealed that C2 rendered non-competitive inhibition and significantly reduced MNase turnover number (Kcat) and catalytic efficiency (Kcat/Km) with an IC50 value of ∼ 1122 nM. In CD spectroscopy, a notable perturbation in the β-sheet content of MNase was observed in presence of C2. Fluorescence-microscope analysis indicated that MNase inhibition by C2 could restore entrapment of methicillin-resistant Staphylococcus aureus (MRSA) in calf-thymus DNA (CT-DNA). Flow cytometry and confocal microscope analysis revealed that uptake of DNA-entrapped MRSA by activated THP-1 cells was reinstated by MNase inhibition rendered by C2. Inhibition of nuclease by the non-toxic ligand C2 holds therapeutic prospect as it has the potential to bolster the DNA-mediated entrapment machinery and mitigate MRSA infections.
Collapse
Affiliation(s)
- Barlina Konwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Sagnik De
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Gopal Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Aiyagari Ramesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
5
|
Ruseva N, Atanasova M, Sbirkova-Dimitrova H, Marković A, Šmelcerović Ž, Šmelcerović A, Cherneva E, Bakalova A. Chloro-substituted pyridine squaramates as new DNase I inhibitors: Synthesis, structural characterization, in vitro evaluation and molecular docking studies. Chem Biol Interact 2023; 386:110772. [PMID: 37898285 DOI: 10.1016/j.cbi.2023.110772] [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: 09/01/2023] [Revised: 10/08/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023]
Abstract
Having continued our recent study on the synthesis and DNase I inhibition of several monosquaramides, two new chloro-substituted pyridine squaramates were synthesized and their structure was identified by X-ray. Their inhibitory properties towards deoxyribonuclease I (DNase I) and xanthine oxidase (XO) were evaluated in vitro. 3-(((6-Chloropyridin-3-yl)methyl)amino)-4-ethoxycyclobut-3-ene-1,2-dione (compound 3a) inhibited DNase I with an IC50 value of 43.82 ± 6.51 μM, thus standing out as one of the most potent small organic DNase I inhibitors tested to date. No cytotoxicity to human tumor cell lines (HL-60, MDA-MB-231 and MCF-7) was observed for the tested compounds. In order to investigate the drug-likeness of the squaramates, the ADME profile and pharmacokinetic properties were evaluated. Molecular docking was performed to reveal the binding mode of the studied compounds on DNase I.
Collapse
Affiliation(s)
- Nina Ruseva
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000, Sofia, Bulgaria
| | - Mariyana Atanasova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000, Sofia, Bulgaria
| | - Hristina Sbirkova-Dimitrova
- Institute of Mineralogy and Crystallography "Akad. Ivan Kostov", Bulgarian Academy of Sciences, Acad. G. Bonchev Bl. 107, 1113, Sofia, Bulgaria
| | - Ana Marković
- Department of Pharmacy, Faculty of Medicine, University of Niš, Bulevar Zorana Ðindića 81, 18000, Niš, Serbia
| | - Žaklina Šmelcerović
- Center for Biomedicinal Science, Faculty of Medicine, University of Niš, Bulevar Zorana Ðindića 81, 18000, Niš, Serbia
| | - Andrija Šmelcerović
- Department of Chemistry, Faculty of Medicine, University of Niš, Bulevar Zorana Ðindića 81, 18000, Niš, Serbia.
| | - Emiliya Cherneva
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000, Sofia, Bulgaria; Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Build. 9, 1113, Sofia, Bulgaria
| | - Adriana Bakalova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000, Sofia, Bulgaria.
| |
Collapse
|
6
|
Islam MM, Sreeharsha N, Alshabrmi FM, Asif AH, Aldhubiab B, Anwer MK, Krishnasamy R, Rehman A. From seeds to survival rates: investigating Linum usitatissimum's potential against ovarian cancer through network pharmacology. Front Pharmacol 2023; 14:1285258. [PMID: 37964873 PMCID: PMC10642394 DOI: 10.3389/fphar.2023.1285258] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/02/2023] [Indexed: 11/16/2023] Open
Abstract
Ovarian cancer is a malignant tumor that primarily forms in the ovaries. It often goes undetected until it has spread to the pelvis and abdomen, making it more challenging to treat and often fatal. Historically, natural products and their structural analogues have played a pivotal role in pharmacotherapy, especially for cancer. Numerous studies have demonstrated the therapeutic potential of Linum usitatissimum against ovarian cancer, but the specific molecular mechanisms remain elusive. This study combines data mining, network pharmacology, and molecular docking analysis to pioneer an innovative approach for ovarian cancer treatment by identifying potent phytochemicals. Findings of current study revealed that Apigenin, Vitamin E, Palmitic acid, Riboflavin, Isolariciresinol, 5-Dehydro-avenasterol, Cholesterol, Pantothenic acid, Nicotinic acid, Campesterol, Beta-Sitosterol, Stigmasterol, Daucosterol, and Vitexin suppress tumor growth by influencing AKT1, JUN, EGFR, and VEGFA. Kaplan-Meier survival analysis spotlighted AKT1, JUN, EGFR, and VEGFA as potential diagnostic and prognostic biomarkers for ovarian cancer. However, it is imperative to conduct in vivo and in vitro examinations to ascertain the pharmacokinetics and biosafety profiles, bolstering the candidacy of L. usitatissimum in ovarian cancer therapeutics.
Collapse
Affiliation(s)
- Mohammed Monirul Islam
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Bangalore, India
| | - Fahad M. Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Afzal Haq Asif
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Bandar Aldhubiab
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, China
| |
Collapse
|
7
|
Aloufi B, Alshabrmi FM, Sreeharsha N, Rehman A. Exploring therapeutic targets and drug candidates for obesity: a combined network pharmacology, bioinformatics approach. J Biomol Struct Dyn 2023; 42:11879-11900. [PMID: 37811763 DOI: 10.1080/07391102.2023.2265491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/24/2023] [Indexed: 10/10/2023]
Abstract
The remarkably high prevalence of obesity in Saudi Arabia reflects a global epidemic demanding urgent attention due to its associated health risks. The integration of traditional medicine, a vital cultural aspect, involves the use of medicinal plants to address various diseases, including obesity. This research merges network pharmacology (NP) and bioinformatics to innovate obesity treatment by identifying effective phytochemicals from native plants in the Taif valley. Focusing on six indigenous plants-Senna alexandrina, Capsicum annuum, Zingiber officinale, Curcuma longa, Trigonella foenum-graecum, and Foeniculum vulgare-we conducted preliminary screenings for potential bioactive compounds. We systematically compiled compound data from public databases and reviewed literature, revealing active compounds like apigenin, kaempferol, moupinamide, cyclocurcumin, chrysoeriol, isorhamnetin, rheinanthrone, cyclocurcumin, and riboflavin.Constructing a compound-target genes-obesity network unveiled their significant impact on metabolic regulation and fat accumulation, interacting notably with key proteins AKT1 and PTGS2. Molecular docking and 100 ns Molecular Dynamic (MD) simulations demonstrated robust binding affinity and stability at the docking site. Employing adipocytes as a cellular model, we gauged their viability and response to obesity-related stressors post-treatment with these native plant compounds.In conclusion, Saudi Arabia's indigenous plants hold promise as natural solutions for obesity treatment. This research opens new avenues in the battle against this pervasive health crisis by incorporating the potential of native botanicals.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Bandar Aloufi
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Bangalore, India
| | - Abdur Rehman
- Department of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
8
|
Konwar B, Mullick P, Das G, Ramesh A. Anthraquinone-Based Ligands as MNase Inhibitors: Insights from Inhibition Studies and Generation of a Payload Nanocarrier for Potential Anti-MRSA Therapy. ChemMedChem 2023; 18:e202200711. [PMID: 37062965 DOI: 10.1002/cmdc.202200711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/14/2023] [Accepted: 04/14/2023] [Indexed: 04/18/2023]
Abstract
The present study highlights the prospect of an anthraquinone-based ligand (C1) as an inhibitor of micrococcal nuclease (MNase) enzyme secreted by Staphylococcus aureus. MNase inhibition rendered by 5.0 μM C1 was ∼96 % and the ligand could significantly distort the β-sheet conformation present in MNase. Mechanistic studies revealed that C1 rendered non-competitive inhibition, reduced the turnover (Kcat ) and catalytic efficiency (Km /Kcat ) of MNase with an IC50 value of 323 nM. C1 could also inhibit nuclease present in the cell-free supernatant (CFS) of a methicillin-resistant Staphylococcus aureus (MRSA) strain. A C1-loaded human serum albumin (HSA)-based nanocarrier (C1-HNC) was developed, which was amicable to protease-triggered release of payload in presence of the CFS of an MRSA strain. Eluates from C1-HNC could effectively reduce the rate of MNase-catalyzed DNA cleavage. The non-toxic nature of C1-HNC in conjunction with the non-competitive mode of MNase inhibition rendered by C1 offers interesting therapeutic prospect in alleviation of MRSA infections.
Collapse
Affiliation(s)
- Barlina Konwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781038, Assam, India
| | - Priya Mullick
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781038, Assam, India
- Present address: Department of Biology, Washington University in St Louis S, t Louis, MO 63130, USA
| | - Gopal Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, 781038, Assam, India
| | - Aiyagari Ramesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781038, Assam, India
| |
Collapse
|
9
|
Ruseva N, Sbirkova-Dimitrova H, Atanasova M, Marković A, Šmelcerović Ž, Šmelcerović A, Bakalova A, Cherneva E. Synthesis and DNase I Inhibitory Properties of New Squaramides. Molecules 2023; 28:molecules28020538. [PMID: 36677597 PMCID: PMC9863136 DOI: 10.3390/molecules28020538] [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: 11/29/2022] [Revised: 12/19/2022] [Accepted: 12/25/2022] [Indexed: 01/09/2023] Open
Abstract
Three new monosquaramides (3a-c) were synthesized, characterized by IR, NMR and X-ray, and evaluated for inhibitory activity against deoxyribonuclease I (DNase I) and xanthine oxidase (XO) in vitro. The target compounds inhibited DNase I with IC50 values below 100 μM, being at the same time more potent DNase I inhibitors than crystal violet, used as a positive control. 3-Ethoxy-4-((1-(pyridin-3-yl)propan-2-yl)amino)cyclobut-3-ene-1,2-dione (3c) stood out as the most potent compound, exhibiting a slightly better IC50 value (48.04 ± 7.98 μM) compared to the other two compounds. In order to analyze potential binding sites for the studied compounds with DNase I, a molecular docking study was performed. Compounds 3a-c are among the most potent small organic DNase I inhibitors tested to date.
Collapse
Affiliation(s)
- Nina Ruseva
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria
| | - Hristina Sbirkova-Dimitrova
- Institute of Mineralogy and Crystallography “Akad. Ivan Kostov”, Bulgarian Academy of Sciences, Acad. G. Bonchev Bl. 107, 1113 Sofia, Bulgaria
| | - Mariyana Atanasova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria
| | - Ana Marković
- Department of Pharmacy, Faculty of Medicine, University of Niš, Bulevar Zorana Đinđića 81, 18000 Niš, Serbia
| | - Žaklina Šmelcerović
- Center for Biomedicinal Science, Faculty of Medicine, University of Niš, Bulevar Zorana Đinđića 81, 18000 Niš, Serbia
| | - Andrija Šmelcerović
- Department of Chemistry, Faculty of Medicine, University of Niš, Bulevar Zorana Đinđića 81, 18000 Niš, Serbia
- Correspondence: (A.Š.); (E.C.)
| | - Adriana Bakalova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria
| | - Emiliya Cherneva
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Build. 9, 1113 Sofia, Bulgaria
- Correspondence: (A.Š.); (E.C.)
| |
Collapse
|
10
|
Kocic G, Hadzi-Djokic J, Colic M, Veljkovic A, Tomovic K, Roumeliotis S, Smelcerovic A, Liakopoulos V. The Role of Nucleases Cleaving TLR3, TLR7/8 and TLR9 Ligands, Dicer RNase and miRNA/piRNA Proteins in Functional Adaptation to the Immune Escape and Xenophagy of Prostate Cancer Tissue. Int J Mol Sci 2022; 24:ijms24010509. [PMID: 36613950 PMCID: PMC9820234 DOI: 10.3390/ijms24010509] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/10/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
The prototypic sensors for the induction of innate and adaptive immune responses are the Toll-like receptors (TLRs). Unusually high expression of TLRs in prostate carcinoma (PC), associated with less differentiated, more aggressive and more propagating forms of PC, changed the previous paradigm about the role of TLRs strictly in immune defense system. Our data reveal an entirely novel role of nucleic acids-sensing Toll-like receptors (NA-TLRs) in functional adaptation of malignant cells for supply and digestion of surrounding metabolic substrates from dead cells as specific mechanism of cancer cells survival, by corresponding ligands accelerated degradation and purine/pyrimidine salvage pathway. The spectrophotometric measurement protocols used for the determination of the activity of RNases and DNase II have been optimized in our laboratory as well as the enzyme-linked immunosorbent method for the determination of NF-κB p65 in prostate tissue samples. The protocols used to determine Dicer RNase, AGO2, TARBP2 and PIWIL4 were based on enzyme-linked immunosorbent assay. The amount of pre-existing acid-soluble oligonucleotides was measured and expressed as coefficient of absorbance. The activities of acid DNase II and RNase T2, and the activities of nucleases cleaving TLR3, TLR7/8 and TLR9 ligands (Poly I:C, poly U and unmethylated CpG), increased several times in PC, compared to the corresponding tumor adjacent and control tissue, exerting very high sensitivity and specificity of above 90%. Consequently higher levels of hypoxanthine and NF-κB p65 were reported in PC, whereas the opposite results were observed for miRNA biogenesis enzyme (Dicer RNase), miRNA processing protein (TARB2), miRNA-induced silencing complex protein (Argonaute-AGO) and PIWI-interacting RNAs silence transposon. Considering the crucial role of purine and pyrimidine nucleotides as energy carriers, subunits of nucleic acids and nucleotide cofactors, future explorations will be aimed to design novel anti-cancer immune strategies based on a specific acid endolysosomal nuclease inhibition.
Collapse
Affiliation(s)
- Gordana Kocic
- Department of Biochemistry, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
- Correspondence: or ; Tel.: +381-63-8122522
| | | | - Miodrag Colic
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| | - Andrej Veljkovic
- Department of Biochemistry, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Katarina Tomovic
- Department of Pharmacy, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Stefanos Roumeliotis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Andrija Smelcerovic
- Department of Chemistry, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Vassilios Liakopoulos
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| |
Collapse
|
11
|
Liu Y, Liang S, Wang B, Zhao J, Zi X, Yan S, Dou T, Jia J, Wang K, Ge C. Advances in Single-Cell Sequencing Technology and Its Application in Poultry Science. Genes (Basel) 2022; 13:genes13122211. [PMID: 36553479 PMCID: PMC9778011 DOI: 10.3390/genes13122211] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Single-cell sequencing (SCS) uses a single cell as the research material and involves three dimensions: genes, phenotypes and cell biological mechanisms. This type of research can locate target cells, analyze the dynamic changes in the target cells and the relationships between the cells, and pinpoint the molecular mechanism of cell formation. Currently, a common problem faced by animal husbandry scientists is how to apply existing science and technology to promote the production of high-quality livestock and poultry products and to breed livestock for disease resistance; this is also a bottleneck for the sustainable development of animal husbandry. In recent years, although SCS technology has been successfully applied in the fields of medicine and bioscience, its application in poultry science has been rarely reported. With the sustainable development of science and technology and the poultry industry, SCS technology has great potential in the application of poultry science (or animal husbandry). Therefore, it is necessary to review the innovation of SCS technology and its application in poultry science. This article summarizes the current main technical methods of SCS and its application in poultry, which can provide potential references for its future applications in precision breeding, disease prevention and control, immunity, and cell identification.
Collapse
Affiliation(s)
- Yong Liu
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Shuangmin Liang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Bo Wang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Jinbo Zhao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Xiannian Zi
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Shixiong Yan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Tengfei Dou
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Junjing Jia
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Kun Wang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Changrong Ge
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Correspondence:
| |
Collapse
|
12
|
Sultana R, Abid OUR, Sultana N, Fakhar-e-Alam M, Siddique MH, Atif M, Nawaz M, Wadood A, Rehman AU, Farooq W, Shafeeq S, Afzal M. Potential Enzyme Inhibitor Triazoles from Aliphatic esters: Synthesis, enzyme inhibition and docking studies. JOURNAL OF SAUDI CHEMICAL SOCIETY 2022. [DOI: 10.1016/j.jscs.2022.101565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
13
|
Garcia Gonzalez J, Hernandez FJ. Nuclease activity: an exploitable biomarker in bacterial infections. Expert Rev Mol Diagn 2022; 22:265-294. [PMID: 35240900 DOI: 10.1080/14737159.2022.2049249] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION In the increasingly challenging field of clinical microbiology, diagnosis is a cornerstone whose accuracy and timing are crucial for the successful management, therapy, and outcome of infectious diseases. Currently employed biomarkers of infectious diseases define the scope and limitations of diagnostic techniques. As such, expanding the biomarker catalog is crucial to address unmet needs and bring about novel diagnostic functionalities and applications. AREAS COVERED This review describes the extracellular nucleases of 15 relevant bacterial pathogens and discusses the potential use of nuclease activity as a diagnostic biomarker. Articles were searched for in PubMed using terms: "nuclease", "bacteria", "nuclease activity" or "biomarker". For overview sections, original and review articles between 2000 and 2019 were searched for using terms: "infections", "diagnosis", "bacterial", "burden", "challenges". Informative articles were selected. EXPERT OPINION Using the catalytic activity of nucleases offers new possibilities compared to established biomarkers. Nucleic acid activatable reporters in combination with different transduction platforms and delivery methods can be used to detect disease-associated nuclease activity patterns in vitro and in vivo for prognostic and diagnostic applications. Even when these patterns are not obvious or of unknown etiology, screening platforms could be used to identify new disease reporters.
Collapse
Affiliation(s)
- Javier Garcia Gonzalez
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.,Wallenberg Centre for Molecular Medicine (WCMM), Linköping, Sweden.,Nucleic Acids Technologies Laboratory (NAT-lab), Linköping University, Linköping, Sweden
| | - Frank J Hernandez
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.,Wallenberg Centre for Molecular Medicine (WCMM), Linköping, Sweden.,Nucleic Acids Technologies Laboratory (NAT-lab), Linköping University, Linköping, Sweden
| |
Collapse
|
14
|
Gajic M, Knez D, Sosič I, Mravljak J, Meden A, Košak U, Leitzbach L, George S, Hofmann B, Zivkovic A, Steinhilber D, Stark H, Gobec S, Smelcerovic A, Anderluh M. Repurposing of 8-Hydroxyquinoline-based Butyrylcholinesterase and Cathepsin B Ligands as Potent Non-peptidic Deoxyribonuclease I Inhibitors. ChemMedChem 2022; 17:e202100694. [PMID: 34994078 DOI: 10.1002/cmdc.202100694] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/20/2021] [Indexed: 11/06/2022]
Abstract
A library of 31 butyrylcholinesterase (BChE) and cathepsin B (CatB) inhibitors, was screened in vitro for inhibition of deoxyribonuclease I (DNase I). Compounds 22, 8 and 7 are among the most potent synthetic non-peptide DNase I inhibitors reported up to date. Three 8-hydroxyquinoline analogues inhibited both DNase I and BChE with IC50 values below 35 µM and 50 nM, respectively, while 2 nitroxoline derivatives inhibited DNase I and Cat B endopeptidase activity with IC50 values below 60 µM and 20 µM, respectively. Selected derivatives were screened for various co-target binding affinities at dopamine D2 and D3, histamine H3 and H4 receptors and inhibition of 5-lipoxygenase. Compound 8 bound to the H3 receptor and is highlighted as the most promising multifunctional ligand with a favorable pharmacokinetic profile and one of the most potent non-peptide DNase I inhibitors. The present study demonstrates that 8-hydroxyquinoline is a structural fragment critical for DNase I inhibition in the presented series of compounds.
Collapse
Affiliation(s)
| | - Damijan Knez
- University of Ljubljana, Department of Medicinal Chemistry, SLOVENIA
| | - Izidor Sosič
- University of Ljubljana, Department of Medicinal Chemistry, SLOVENIA
| | - Janez Mravljak
- University of Ljubljana, Department of Medicinal Chemistry, SLOVENIA
| | - Anže Meden
- University of Ljubljana, Department of Medicinal Chemistry, SLOVENIA
| | - Urban Košak
- University of Ljubljana, Department of Medicinal Chemistry, SLOVENIA
| | - Luisa Leitzbach
- Heinrich Heine University Duesseldorf, Institute for Pharmaceutical and Medicinal Chemistry, GERMANY
| | - Sven George
- Goethe-University of Frankfurt, Institute of Pharmaceutical Chemistry, GERMANY
| | - Bettina Hofmann
- Goethe-University of Frankfurt, Institute of Pharmaceutical Chemistry, GERMANY
| | - Aleksandra Zivkovic
- Heinrich Heine University Duesseldorf, Institute of Pharmaceutical and Medicinal Chemistry, GERMANY
| | - Dieter Steinhilber
- Goethe-University of Frankfurt, Institute of Pharmaceutical Chemistry, GERMANY
| | - Holger Stark
- Heinrich Heine University Duesseldorf, Institute of Pharmaceutical and Medicinal Chemistry, GERMANY
| | - Stanislav Gobec
- University of Ljubljana, Department of Medicinal Chemistry, SLOVENIA
| | | | - Marko Anderluh
- University of Ljubljana, Faculty of Pharmacy, Askerceva cesta 7, 1000, Ljubljana, SLOVENIA
| |
Collapse
|
15
|
DNase activity in human seminal plasma and follicular fluid and its inhibition by follicular fluid chelating agents. Reprod Biomed Online 2021; 43:1079-1086. [PMID: 34753679 DOI: 10.1016/j.rbmo.2021.09.015] [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: 05/17/2021] [Revised: 07/30/2021] [Accepted: 09/20/2021] [Indexed: 11/23/2022]
Abstract
RESEARCH QUESTION What is the mechanism by which human follicular fluid inhibits seminal plasma DNase activity? DESIGN Human genomic DNA was incubated with human follicular fluid and seminal plasma (reaction mixture) under different experimental conditions; increasing volumes of human follicular fluid; proteinase K digested or heat inactivated human follicular fluid; and the addition of Ca2+ or Mg2+ to the reaction mixture. RESULTS Increasing volume of human follicular fluid resulted in a dose-dependent inhibition of seminal plasma DNase activity. Inhibition was not caused by proteins in the human follicular fluid as digestion with proteinase K or heat inactivation of human follicular fluid failed to abolish its inhibitory effect. Addition of divalent cations resulted in a reversion of the inhibitory effect, providing evidence that human follicular fluid inhibition of seminal plasma DNase activity seems to be mediated by a compound with chelating activity. Furthermore, incubation of genomic DNA with human follicular fluid in the presence of divalent cations served to elicit the existence of DNase activity. CONCLUSIONS Human follicular fluid seems to contain a molecule or molecules with chelating capacity that inhibits DNase activity of both follicular fluid and seminal plasma. Our findings provide new insight to understanding sperm preservation and the physiology of fertilization biology.
Collapse
|
16
|
Gajić M, Ilić BS, Bondžić BP, Džambaski Z, Kojić VV, Jakimov DS, Kocić G, Šmelcerović A. 1,2,3,4-Tetrahydroisoquinoline Derivatives as a Novel Deoxyribonuclease I Inhibitors. Chem Biodivers 2021; 18:e2100261. [PMID: 34170076 DOI: 10.1002/cbdv.202100261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022]
Abstract
Herein we report an assessment of 24 1,2,3,4-tetrahydroisoquinoline derivatives for potential DNase I (deoxyribonuclease I) inhibitory properties in vitro. Four of them inhibited DNase I with IC50 values below 200 μM. The most potent was 1-(6,7-dimethoxy-1,2,3,4-tetrahydroisoquinolin-1-yl)propan-2-one (2) (IC50 =134.35±11.38 μM) exhibiting slightly better IC50 value compared to three other active compounds, 2-[2-(4-fluorophenyl)-1,2,3,4-tetrahydroisoquinolin-1-yl]-1-phenylethan-1-one (15) (IC50 =147.51±14.87 μM), 2-[2-(4-fluorophenyl)-1,2,3,4-tetrahydroisoquinolin-1-yl]cyclohexan-1-one (18) (IC50 =149.07±2.98 μM) and 2-[6,7-dimethoxy-2-(p-tolyl)-1,2,3,4-tetrahydroisoquinolin-1-yl]cyclohexan-1-one (22) (IC50 =148.31±2.96 μM). Cytotoxicity assessment of the active DNase I inhibitors revealed a lack of toxic effects on the healthy cell lines MRC-5. Molecular docking and molecular dynamics simulations suggest that interactions with Glu 39, His 134, Asn 170, Tyr 211, Asp 251 and His 252 are an important factor for inhibitors affinity toward the DNase I. Observed interactions would be beneficial for the discovery of new active 1,2,3,4-tetrahydroisoquinoline-based inhibitors of DNase I, but might also encourage researchers to further explore and utilize potential therapeutic application of DNase I inhibitors, based on a versatile role of DNase I during apoptotic cell death.
Collapse
Affiliation(s)
- Mihajlo Gajić
- University of Niš, Faculty of Medicine, Department of Pharmacy, Blvd. Dr. Zorana Đinđića 81, 18000, Niš, Serbia
| | - Budimir S Ilić
- University of Niš, Faculty of Medicine, Department of Chemistry, Blvd. Dr. Zorana Đinđića 81, 18000, Niš, Serbia
| | - Bojan P Bondžić
- University of Belgrade, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, Njegoševa 12, 11000, Belgrade, Serbia
| | - Zdravko Džambaski
- University of Belgrade, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, Njegoševa 12, 11000, Belgrade, Serbia
| | - Vesna V Kojić
- University of Novi Sad, Faculty of Medicine, Oncology Institute of Vojvodina, Put Dr. Goldmana 4, 21204, Sremska Kamenica, Serbia
| | - Dimitar S Jakimov
- University of Novi Sad, Faculty of Medicine, Oncology Institute of Vojvodina, Put Dr. Goldmana 4, 21204, Sremska Kamenica, Serbia
| | - Gordana Kocić
- University of Niš, Faculty of Medicine, Department of Biochemistry, Blvd. Dr. Zorana Đinđića 81, 18000, Niš, Serbia
| | - Andrija Šmelcerović
- University of Niš, Faculty of Medicine, Department of Chemistry, Blvd. Dr. Zorana Đinđića 81, 18000, Niš, Serbia
| |
Collapse
|
17
|
Gajić M, Džambaski Z, Ilić BS, Kocić G, Bondžić BP, Šmelcerović A. Synthesis and analysis of 4-oxothiazolidines as potential dual inhibitors of deoxyribonuclease I and xanthine oxidase. Chem Biol Interact 2021; 345:109536. [PMID: 34058176 DOI: 10.1016/j.cbi.2021.109536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/16/2021] [Accepted: 05/24/2021] [Indexed: 10/21/2022]
Abstract
In this study, seven new 4-oxothiazolidine derivatives were synthesized and assayed, along 7 known derivatives, for inhibitory properties against deoxyribonuclease I (DNase I) and xanthine oxidase (XO) in vitro. Among tested compounds, (5Z)-Ethyl-2-(2-(cyanomethylene)-4-oxothiazolidin-5-yliden)acetate (6) exhibited inhibitory activity against both enzymes (DNase I IC50 = 67.94 ± 5.99 μM; XO IC50 = 98.98 ± 13.47 μM), therefore being the first reported dual inhibitor of DNase I and XO. Observed DNase I inhibition qualifies compound 6 as the most potent small organic DNase I inhibitor reported so far. Derivatives of 2-alkyliden-4-oxothiazolidinone (1) inhibited DNase I below 200 μM, while the other tested 4-oxothiazolidine derivatives remained inactive against both enzymes. The molecular docking and molecular dynamics simulations into the binding sites of DNase I and XO enzyme allowed us to clarify the binding modes of this 4-oxothiazolidine derivative, which might aid future development of dual DNase I and XO.
Collapse
Affiliation(s)
- Mihajlo Gajić
- University of Niš, Faculty of Medicine, Department of Pharmacy, Blvd. Dr. Zorana Đinđića 81, 18000, Niš, Serbia
| | - Zdravko Džambaski
- University of Belgrade, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, Njegoševa 12, 11000, Belgrade, Serbia
| | - Budimir S Ilić
- University of Niš, Faculty of Medicine, Department of Chemistry, Blvd. Dr. Zorana Đinđića 81, 18000, Niš, Serbia
| | - Gordana Kocić
- University of Niš, Faculty of Medicine, Department of Biochemistry, Blvd. Dr. Zorana Đinđića 81, 18000, Niš, Serbia
| | - Bojan P Bondžić
- University of Belgrade, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, Njegoševa 12, 11000, Belgrade, Serbia.
| | - Andrija Šmelcerović
- University of Niš, Faculty of Medicine, Department of Chemistry, Blvd. Dr. Zorana Đinđića 81, 18000, Niš, Serbia.
| |
Collapse
|
18
|
Ilić BS, Gajić M, Bondžić BP, Džambaski Z, Kocić G, Šmelcerović A. Deoxyribonuclease I Inhibitory Properties, Molecular Docking and Molecular Dynamics Simulations of 1‐(Pyrrolidin‐2‐yl)propan‐2‐one Derivatives. Chem Biodivers 2021; 18:e2000996. [DOI: 10.1002/cbdv.202000996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/02/2021] [Indexed: 12/20/2022]
Affiliation(s)
- Budimir S. Ilić
- University of Niš Faculty of Medicine, Department of Chemistry Blvd. Dr. Zorana Đinđića 81 18000 Niš Serbia
| | - Mihajlo Gajić
- University of Niš Faculty of Medicine, Department of Pharmacy Blvd. Dr. Zorana Đinđića 81 18000 Niš Serbia
| | - Bojan P. Bondžić
- University of Belgrade Institute of Chemistry, Technology and Metallurgy Njegoševa 12 11000 Belgrade Serbia
| | - Zdravko Džambaski
- University of Belgrade Institute of Chemistry, Technology and Metallurgy Njegoševa 12 11000 Belgrade Serbia
| | - Gordana Kocić
- University of Niš Faculty of Medicine, Department of Biochemistry Blvd. Dr. Zorana Đinđića 81 18000 Niš Serbia
| | - Andrija Šmelcerović
- University of Niš Faculty of Medicine, Department of Chemistry Blvd. Dr. Zorana Đinđića 81 18000 Niš Serbia
| |
Collapse
|
19
|
Abstract
Donor-derived cell-free DNA (dd-cfDNA) in the blood circulation is an early marker of injury in solid organ transplantation. Here, we review recent evidence that indicates that dd-cfDNA may itself be a trigger of inflammation, thereby adding insult on injury. Early unresolving molecular allograft injury measured via changes in dd-cfDNA may be an early warning sign and may therefore enable stratification of patients who are at risk of subsequent allograft injury. Considering dd-cfDNA as a continuous and clinically significant biomarker opens up the potential for new management strategies, therapeutics, and ways to quantify interventions by considering the immunological potential of dd-cfDNA.
Collapse
|
20
|
Interplay between male reproductive system dysfunction and the therapeutic effect of flavonoids. Fitoterapia 2020; 147:104756. [PMID: 33069836 DOI: 10.1016/j.fitote.2020.104756] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 12/20/2022]
Abstract
Male infertility has affected many families around the world. However, due to the mechanism underlying male reproductive system dysfunction are not completely elucidated, the use of drugs for male reproductive system dysfunction treatment only insignificant higher pregnancy outcomes, low-quality evidence suggests that clinical pregnancy rates may increase. Therefore, the focus in the future will be on developing more viable treatment options to prevent or treatment of male reproductive system dysfunction and achieve the purpose of improving fertility. Interestingly, natural products, as the potential inhibitors for the treatment of male reproductive system dysfunction, have shown a good therapeutic effect. Among many natural products, flavonoids have been extensively investigated for the treatment of male reproductive system dysfunction, such as testicular structural disruption, spermatogenesis disturbance and sperm quality decline. Flavonoids have been reported to have antioxidant, anti-inflammatory, immune stimulating, anti-apoptotic, anticarcinogenic, anti-allergic and antiviral activities, investigating for the treatment of male reproductive system dysfunction. In this review, we evaluate the therapeutic effects of flavonoids on male reproductive system dysfunction under different cellular scenarios and summarize the therapeutic strategies of flavonoids based on the aforementioned retrospective analysis. In the end, we describe some perspective research areas relevant to the application of flavonoids in the treatment of male reproductive system dysfunction.
Collapse
|
21
|
Wilson-Robles H, Miller T, Jarvis J, Terrell J, Dewsbury N, Kelly T, Herzog M, Bygott T, Hardat N, Michel G. Evaluation of nucleosome concentrations in healthy dogs and dogs with cancer. PLoS One 2020; 15:e0236228. [PMID: 32866177 PMCID: PMC7458307 DOI: 10.1371/journal.pone.0236228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/12/2020] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Nucleosomes consist of small fragments of DNA wrapped around a histone octamer core. Diseases such as cancer or inflammation lead to cell death, which causes fragmentation and release of nucleosomes into the blood. The Nu.Q™ technology measures circulating nucleosome levels and exploits the different compositions of cancer derived nucleosomes in blood to detect and identify cancer even at early stages. The objectives of this study are to identify the optimal sample type for the Nu.Q™ H3.1 assay and to determine if it can accurately detect nucleosomes in the blood of healthy canines as well as those with cancer. MATERIALS AND METHODS Blood samples from healthy canine volunteers as well as dogs newly diagnosed with lymphoma were used. The blood was processed at a variety of times under a variety of conditions to determine the most reliable sample type and conditions, and to develop an appropriate processing strategy to ensure reliably accurate results. RESULTS Nucleosomes could be detected using a variety of sample collection and processing protocols. Nucleosome signals were highest in EDTA plasma and serum samples and most consistent in plasma. Samples should be processed within an hour of collection. Experiments showed that samples were able to withstand several freeze thaw cycles. Processing time and tcollection tube type did affect nucleosome detection levels. Finally, significantly elevated concentrations of nucleosomes were seen in a small cohort of dogs that had been newly diagnosed with lymphoma. CONCLUSIONS When samples are collected and processed appropriately, the Nu.Q™ platform can reliably detect nucleosomes in the plasma of dogs. Further testing is underway to validate and optimize the Nu.Q™ platform for veterinary use.
Collapse
Affiliation(s)
- Heather Wilson-Robles
- Small Animal Clinical Sciences Department, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
- * E-mail:
| | - Tasha Miller
- Small Animal Clinical Sciences Department, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Jill Jarvis
- Small Animal Clinical Sciences Department, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Jason Terrell
- Volition America & Volition Veterinary Diagnostic Development, Austin, Texas, United States of America
| | - Nathan Dewsbury
- Volition America & Volition Veterinary Diagnostic Development, Austin, Texas, United States of America
| | - Terry Kelly
- Volition America & Volition Veterinary Diagnostic Development, Austin, Texas, United States of America
| | | | | | | | | |
Collapse
|
22
|
Zhao M, Huang S, Xie H, Wang J, Zhao X, Li M, Zhao M. Construction of Specific and Reversible Nanoreceptors for Proteins via Sequential Surface-Imprinting Strategy. Anal Chem 2020; 92:10540-10547. [DOI: 10.1021/acs.analchem.0c01366] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Muhua Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P. R. China
| | - Shan Huang
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P. R. China
| | - Huaisyuan Xie
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P. R. China
| | - Jiayu Wang
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P. R. China
| | - Xiaoli Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P. R. China
| | - Mengyuan Li
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P. R. China
| | - Meiping Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P. R. China
| |
Collapse
|
23
|
Inhibition of nuclease activity by a splice-switching oligonucleotide targeting deoxyribonuclease 1 mRNA prevents apoptosis progression and prolong viability of normal human CD4 + T-lymphocytes. Biochimie 2020; 174:34-43. [PMID: 32315661 DOI: 10.1016/j.biochi.2020.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/27/2020] [Accepted: 04/10/2020] [Indexed: 11/21/2022]
Abstract
The nuclease activity of deoxyribonuclease 1 (DNase I) is regulated by alternative splicing (AS) of its mRNA. The aim of this study was to define the ability of a splice-switching oligonucleotide (SSO) that base-paired with DNase I pre-mRNA to induce AS and inhibit nuclease activity in human T, B and NK lymphocytes. The SSO for DNase I could significantly downregulate the expression of full-length active DNase I and upregulate a truncated splice variant with a deleted exon 4. Such an induction of AS resulted in inhibition of nuclease activity and slowed apoptosis progression in anti-CD95/FAS stimulated lymphocytes. These results should facilitate further investigations of apoptosis regulation in lymphocytes and demonstrate that SSOs for DNase I are promising cytoprotective agents.
Collapse
|
24
|
Zhang J, Li Q, Jiang X, Li X, Dong P, Li J, Komiyama M, Liang X. Effect of sulfated polysaccharides on the digestion of DNA by pepsin under simulated gastric juice in vitro. Food Funct 2020; 11:1790-1797. [PMID: 32053124 DOI: 10.1039/c9fo02578b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The effect of sulfated polysaccharides on the digestion of dietary DNA by pepsin was studied using in vitro simulated gastric juice. The results showed that fucoidan (FUC), dextran sulfate (DS) and chondroitin sulfate (CS) could inhibit the digestion of DNA in a dose-dependent manner. Polysaccharides with high sulfate group content have stronger inhibition ability. Fluorescence spectroscopy results showed that polysaccharides could bind to pepsin, and transmission electron microscopy (TEM) confirmed that polysaccharides can interact with DNA, which not only is the main reason that polysaccharides inhibit the digestion of DNA by pepsin but also causes the digestion of DNA by DNase II to be inhibited. The finding suggests that the digestion of DNA should be reevaluated when eating foods rich in sulfated polysaccharides. This study enriched the known pharmacological properties of sulfated polysaccharides as pepsin inhibitors and provided inspiration for the use of sulfated polysaccharides as oligonucleotide drug delivery carriers.
Collapse
Affiliation(s)
- Jing Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Espinoza-Culupú A, Mendes E, Vitorino HA, da Silva PI, Borges MM. Mygalin: An Acylpolyamine With Bactericidal Activity. Front Microbiol 2020; 10:2928. [PMID: 31998255 PMCID: PMC6965172 DOI: 10.3389/fmicb.2019.02928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/05/2019] [Indexed: 01/15/2023] Open
Abstract
Inappropriate use of antibiotics favors the selection and spread of resistant bacteria. To reduce the spread of these bacteria, finding new molecules with activity is urgent and necessary. Several polyamine analogs have been constructed and used to control microorganisms and tumor cells. Mygalin is a synthetic acylpolyamine, which are analogs of spermidine, derived from the hemolymph of the spider Acanthoscurria gomesiana. The effective activity of polyamines and their analogs has been associated with their structure. The presence of two acyl groups in the Mygalin structure may give this molecule a specific antibacterial activity. The aim of this study was to identify the mechanisms involved in the interaction of Mygalin with Escherichia coli to clarify its antimicrobial action. The results indicated that Mygalin exhibits intense dose and time-dependent bactericidal activity. Treatment of E. coli with this molecule caused membrane rupture, inhibition of DNA synthesis, DNA damage, and morphological changes. The esterase activity increased along with the intracellular production of reactive oxygen species (ROS) after treatment of the bacteria with Mygalin. In addition, this molecule was able to sequester iron and bind to LPS. We have shown that Mygalin has bactericidal activity with underlying mechanisms involving ROS generation and chelation of iron ions that are necessary for bacterial metabolism, which may contribute to its microbicidal activity. Taken together, our data suggest that Mygalin can be explored as a new alternative drug with antimicrobial potential against Gram-negative bacteria or other infectious agents.
Collapse
Affiliation(s)
- Abraham Espinoza-Culupú
- Ph.D. Program in Biotechnology, University of São Paulo, São Paulo, Brazil.,Bacteriology Laboratory, Butantan Institute, São Paulo, Brazil
| | | | - Hector Aguilar Vitorino
- Institute of Marine and Environmental Technology, University of Maryland Center for Environmental Science, Columbus Center, Baltimore, MD, United States
| | | | | |
Collapse
|
26
|
Smelcerovic A, Zivkovic A, Ilic BS, Kolarevic A, Hofmann B, Steinhilber D, Stark H. 4-(4-Chlorophenyl)thiazol-2-amines as pioneers of potential neurodegenerative therapeutics with anti-inflammatory properties based on dual DNase I and 5-LO inhibition. Bioorg Chem 2020; 95:103528. [DOI: 10.1016/j.bioorg.2019.103528] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 11/25/2022]
|
27
|
Gabr MT, Pigge FC. Expanding the Toolbox for Label-Free Enzyme Assays: A Dinuclear Platinum(II) Complex/DNA Ensemble with Switchable Near-IR Emission. Molecules 2019; 24:E4390. [PMID: 31805648 PMCID: PMC6930566 DOI: 10.3390/molecules24234390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022] Open
Abstract
Switchable luminescent bioprobes whose emission can be turned on as a function of specific enzymatic activity are emerging as important tools in chemical biology. We report a promising platform for the development of label-free and continuous enzymatic assays in high-throughput mode based on the reversible solvent-induced self-assembly of a neutral dinuclear Pt(II) complex. To demonstrate the utility of this strategy, the switchable luminescence of a dinuclear Pt(II) complex was utilized in developing an experimentally simple, fast (10 min), low cost, and label-free turn-on luminescence assay for the endonuclease enzyme DNAse I. The complex displays a near-IR (NIR) aggregation-induced emission at 785 nm in aqueous solution that is completely quenched upon binding to G-quadruplex DNA from the human c-myc oncogene. Luminescence is restored upon DNA degradation elicited by exposure to DNAse I. Correlation between near-IR luminescence intensity and DNAse I concentration in human serum samples allows for fast and label-free detection of DNAse I down to 0.002 U/mL. The Pt(II) complex/DNA assembly is also effective for identification of DNAse I inhibitors, and assays can be performed in multiwell plates compatible with high-throughput screening. The combination of sensitivity, speed, convenience, and cost render this method superior to all other reported luminescence-based DNAse I assays. The versatile response of the Pt(II) complex to DNA structures promises broad potential applications in developing real-time and label-free assays for other nucleases as well as enzymes that regulate DNA topology.
Collapse
|
28
|
Synthesis and DNase I inhibitory properties of new benzocyclobutane-2,5-diones. Future Med Chem 2019; 11:2415-2426. [DOI: 10.4155/fmc-2019-0032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Eight new benzocyclobutane-2,5-diones (1a–1h) were synthesized, and their inhibitory properties against bovine pancreatic DNase I were examined in vitro. Methods & results: Compounds 1a–1h were synthesized using photocycloaddition of duroquinone with various phenyl-substituted ethylenes in the presence of 18W compact fluorescent lamp (visible light). Two compounds, 1,3,4,6-tetramethyl-7-phenylbicyclo[4.2.0]oct-3-ene-2,5-dione (1a) and 1,3,4,6-tetramethyl-7-p-tolylbicyclo[4.2.0]oct-3-ene-2,5-dione (1b) inhibited DNase I in a noncompetitive manner with IC50 values below 150 μM and showed to be more potent DNase I inhibitors than crystal violet, used as a positive control. In order to analyze potential binding sites for the studied compounds with DNase I, molecular docking study was performed. Conclusion: The studied benzocyclobutane-2,5-diones offer a good starting point for a design of new DNase I inhibitors.
Collapse
|
29
|
Sex, Age, and Bodyweight as Determinants of Extracellular DNA in the Plasma of Mice: A Cross-Sectional Study. Int J Mol Sci 2019; 20:ijms20174163. [PMID: 31454899 PMCID: PMC6747214 DOI: 10.3390/ijms20174163] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 01/24/2023] Open
Abstract
Extracellular DNA (ecDNA) is studied as a possible biomarker, but also as a trigger of the immune responses important for the pathogenesis of several diseases. Extracellular deoxyribonuclease (DNase) activity cleaves ecDNA. The aim of our study was to describe the interindividual variability of ecDNA and DNase activity in the plasma of healthy mice, and to analyze the potential determinants of the variability, including sex, age, and bodyweight. In this experiment, 58 adult CD1 mice (41 females and 31 males) of a variable age (3 to 16 months old) and bodyweight (females 25.7 to 52.1 g, males 24.6 to 49.6 g) were used. The plasma ecDNA was measured using a fluorometric method. The nuclear ecDNA and mitochondrial ecDNA were quantified using real-time PCR. The deoxyribonuclease activity was assessed using the single radial enzyme diffusion method. The coefficient of variance for plasma ecDNA was 139%, and for DNase 48%. Sex differences were not found in the plasma ecDNA (52.7 ± 73.0 ηg/mL), but in the DNase activity (74.5 ± 33.5 K.u./mL for males, and 47.0 ± 15.4 K.u./mL for females). There were no associations between plasma ecDNA and bodyweight or the age of mice. Our study shows that the variability of plasma ecDNA and DNase in adult healthy mice is very high. Sex, age, and bodyweight seem not to be major determinants of ecDNA variability in healthy mice. As ecDNA gains importance in the research of several diseases, it is of importance to understand its production and cleavage. Further studies should, thus, test other potential determinants, taking into account cleavage mechanisms other than DNase.
Collapse
|
30
|
Kolarevic A, Pavlovic A, Djordjevic A, Lazarevic J, Savic S, Kocic G, Anderluh M, Smelcerovic A. Rutin as Deoxyribonuclease I Inhibitor. Chem Biodivers 2019; 16:e1900069. [DOI: 10.1002/cbdv.201900069] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 03/18/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Ana Kolarevic
- Department of Pharmacy, Faculty of MedicineUniversity of Nis Bulevar Dr Zorana Djindjica 81 18000 Nis Serbia
| | - Aleksandra Pavlovic
- Department of Chemistry, Faculty of Science and MathematicsUniversity of Nis Visegradska 33 18000 Nis Serbia
| | - Aleksandra Djordjevic
- Department of Chemistry, Faculty of Science and MathematicsUniversity of Nis Visegradska 33 18000 Nis Serbia
| | - Jelena Lazarevic
- Department of Chemistry, Faculty of MedicineUniversity of Nis Bulevar Dr Zorana Djindjica 81 18000 Nis Serbia
| | - Sasa Savic
- Faculty of TechnologyUniversity of Nis Bulevar Oslobodjenja 124 16000 Leskovac Serbia
| | - Gordana Kocic
- Institute of Biochemistry, Faculty of MedicineUniversity of Nis Bulevar Dr Zorana Djindjica 81 18000 Nis Serbia
| | - Marko Anderluh
- Department of Pharmaceutical Chemistry, Faculty of PharmacyUniversity of Ljubljana Askerceva cesta 7 1000 Ljubljana Slovenia
| | - Andrija Smelcerovic
- Department of Chemistry, Faculty of MedicineUniversity of Nis Bulevar Dr Zorana Djindjica 81 18000 Nis Serbia
| |
Collapse
|
31
|
GÜVELİ Ş. Inhibition of DNase I Enzyme with Nickel(II) Triphenylphosphine Complexes Incorporating Tridentate Schiff Base Ligands in Vitro. JOURNAL OF THE TURKISH CHEMICAL SOCIETY, SECTION A: CHEMISTRY 2018. [DOI: 10.18596/jotcsa.472530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
32
|
Reichard A, Asosingh K. Best Practices for Preparing a Single Cell Suspension from Solid Tissues for Flow Cytometry. Cytometry A 2018; 95:219-226. [PMID: 30523671 DOI: 10.1002/cyto.a.23690] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/26/2018] [Accepted: 11/02/2018] [Indexed: 01/02/2023]
Abstract
Preparing a single cell suspension is a critical step in any solid tissue flow cytometry experiment. Tissue dissection, enzymatic digestion, and mechanical dissociation are three significant steps leading to the degradation of the extracellular matrix and the isolation of single cells, allowing the generation of high-quality flow cytometry data. Cells and the extracellular matrix contain various proteins and other structures which must be considered when designing a tissue digestion protocol to preserve the viability of cells and the presence of relevant antigens while digesting matrix components and cleaving cell-cell junctions. Evaluation of the single cell suspension is essential before proceeding with the labeling of the cells as high viability and absence of cell debris and aggregates are critical for flow cytometry. The information presented should be used as a general guide of steps to consider when preparing a single cell suspension from solid tissues for flow cytometry experiments. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Andrew Reichard
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Kewal Asosingh
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio.,Flow Cytometry Core, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
33
|
Shen W, Guo K, Adkins GB, Jiang Q, Liu Y, Sedano S, Duan Y, Yan W, Wang SE, Bergersen K, Worth D, Wilson EH, Zhong W. A Single Extracellular Vesicle (EV) Flow Cytometry Approach to Reveal EV Heterogeneity. Angew Chem Int Ed Engl 2018; 57:15675-15680. [PMID: 30291794 PMCID: PMC6246790 DOI: 10.1002/anie.201806901] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/02/2018] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) actively participate in intercellular communication and pathological processes. Studying the molecular signatures of EVs is key to reveal their biological functions and clinical values, which, however, is greatly hindered by their sub-100 nm dimensions, the low quantities of biomolecules each EV carries, and the large population heterogeneity. Now, single-EV flow cytometry analysis is introduced to realize single EV counting and phenotyping in a conventional flow cytometer for the first time, enabled by target-initiated engineering (TIE) of DNA nanostructures on each EV. By illuminating multiple markers on single EVs, statistically significant differences are revealed among the molecular signatures of EVs originating from several breast cancer cell lines, and the cancer cell-derived EVs among the heterogeneous EV populations are successfully recognized. Thus, our approach holds great potential for various biological and biomedical applications.
Collapse
Affiliation(s)
- Wen Shen
- University of California - Riverside, Department of Chemistry, Riverside, CA, 92521, U.S.A.
| | - Kaizhu Guo
- University of California - Riverside, Department of Chemistry, Riverside, CA, 92521, U.S.A.
| | - Gary Brent Adkins
- University of California - Riverside, Department of Chemistry, Riverside, CA, 92521, U.S.A.
| | - Qiaoshi Jiang
- University of California - Riverside, Environmental Toxicology Program, Riverside, CA, 92521, U.S.A
| | - Yang Liu
- University of California - Riverside, Environmental Toxicology Program, Riverside, CA, 92521, U.S.A
| | - Sabrina Sedano
- University of California - Riverside, Department of Chemistry, Riverside, CA, 92521, U.S.A.
| | - Yaokai Duan
- University of California - Riverside, Department of Chemistry, Riverside, CA, 92521, U.S.A.
| | - Wei Yan
- University of California - San Diego, Department of Pathology, La Jolla, CA, 92093, U.S.A
| | - Shizhen Emily Wang
- University of California - San Diego, Department of Pathology, La Jolla, CA, 92093, U.S.A
| | - Kristina Bergersen
- University of California - Riverside, Division of Biomedical Sciences, Riverside, CA, 92521, U.S.A
| | - Danielle Worth
- University of California - Riverside, Division of Biomedical Sciences, Riverside, CA, 92521, U.S.A
| | - Emma H. Wilson
- University of California - Riverside, Division of Biomedical Sciences, Riverside, CA, 92521, U.S.A
| | - Wenwan Zhong
- University of California - Riverside, Department of Chemistry, Riverside, CA, 92521, U.S.A.
- University of California - Riverside, Environmental Toxicology Program, Riverside, CA, 92521, U.S.A
| |
Collapse
|
34
|
Shen W, Guo K, Adkins GB, Jiang Q, Liu Y, Sedano S, Duan Y, Yan W, Wang SE, Bergersen K, Worth D, Wilson EH, Zhong W. A Single Extracellular Vesicle (EV) Flow Cytometry Approach to Reveal EV Heterogeneity. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201806901] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Wen Shen
- University of California-Riverside; Department of Chemistry; Riverside CA 92521 USA
| | - Kaizhu Guo
- University of California-Riverside; Department of Chemistry; Riverside CA 92521 USA
| | - Gary Brent Adkins
- University of California-Riverside; Department of Chemistry; Riverside CA 92521 USA
| | - Qiaoshi Jiang
- University of California-Riverside; Environmental Toxicology Program; Riverside CA 92521 USA
| | - Yang Liu
- University of California-Riverside; Environmental Toxicology Program; Riverside CA 92521 USA
| | - Sabrina Sedano
- University of California-Riverside; Department of Chemistry; Riverside CA 92521 USA
| | - Yaokai Duan
- University of California-Riverside; Department of Chemistry; Riverside CA 92521 USA
| | - Wei Yan
- University of California-San Diego; Department of Pathology; La Jolla CA 92093 USA
| | - Shizhen Emily Wang
- University of California-San Diego; Department of Pathology; La Jolla CA 92093 USA
| | - Kristina Bergersen
- University of California-Riverside; Division of Biomedical Sciences; Riverside CA 92521 USA
| | - Danielle Worth
- University of California-Riverside; Division of Biomedical Sciences; Riverside CA 92521 USA
| | - Emma H. Wilson
- University of California-Riverside; Division of Biomedical Sciences; Riverside CA 92521 USA
| | - Wenwan Zhong
- University of California-Riverside; Department of Chemistry; Riverside CA 92521 USA
- University of California-Riverside; Environmental Toxicology Program; Riverside CA 92521 USA
| |
Collapse
|
35
|
Synthesis and DNase I inhibitory properties of some 5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidines. Bioorg Chem 2018; 80:693-705. [DOI: 10.1016/j.bioorg.2018.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/08/2018] [Accepted: 07/09/2018] [Indexed: 12/11/2022]
|
36
|
Kolarević A, Ilić BS, Kocić G, Džambaski Z, Šmelcerović A, Bondžić BP. Synthesis and DNase I inhibitory properties of some 4‐thiazolidinone derivatives. J Cell Biochem 2018; 120:264-274. [DOI: 10.1002/jcb.27339] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 06/27/2018] [Indexed: 01/10/2023]
Affiliation(s)
- Ana Kolarević
- Department of Pharmacy Faculty of Medicine, University of Niš Niš Serbia
| | - Budimir S. Ilić
- Department of Chemistry Faculty of Medicine, University of Niš Niš Serbia
| | - Gordana Kocić
- Department of Biochemistry, Faculty of Medicine, University of Niš Niš Serbia
| | | | | | - Bojan P. Bondžić
- Center for Chemistry ICTM, University of Belgrade Belgrade Serbia
| |
Collapse
|
37
|
Kolarević A, Ilić BS, Anastassova N, Mavrova AT, Yancheva D, Kocić G, Šmelcerović A. Benzimidazoles as novel deoxyribonuclease I inhibitors. J Cell Biochem 2018; 119:8937-8948. [DOI: 10.1002/jcb.27147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 05/18/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Ana Kolarević
- Department of Pharmacy Faculty of Medicine, University of Niš Niš Serbia
| | - Budimir S. Ilić
- Department of Chemistry Faculty of Medicine, University of Niš Niš Serbia
| | - Neda Anastassova
- 3Laboratory of Structural Organic Analysis, Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences Sofia Bulgaria
| | - Anelia Ts. Mavrova
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy Sofia Bulgaria
| | - Denitsa Yancheva
- 3Laboratory of Structural Organic Analysis, Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences Sofia Bulgaria
| | - Gordana Kocić
- Department of Biochemistry, Faculty of Medicine, University of Niš Niš Serbia
| | | |
Collapse
|
38
|
Sahareen T, Dey P, Mukherjee S, Das G, Ramesh A. Potential of Pyridine Amphiphiles as Staphylococcal Nuclease Inhibitor. Chembiochem 2018; 19:1400-1408. [DOI: 10.1002/cbic.201800032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Tabassum Sahareen
- Department of Biosciences and Bioengineering; Indian Institute of Technology Guwahati; Guwahati Assam 781 039 India
| | - Poulomi Dey
- Department of Biosciences and Bioengineering; Indian Institute of Technology Guwahati; Guwahati Assam 781 039 India
| | - Sandipan Mukherjee
- Department of Biosciences and Bioengineering; Indian Institute of Technology Guwahati; Guwahati Assam 781 039 India
| | - Gopal Das
- Department of Chemistry; Indian Institute of Technology Guwahati; Guwahati Assam 781 039 India
| | - Aiyagari Ramesh
- Department of Biosciences and Bioengineering; Indian Institute of Technology Guwahati; Guwahati Assam 781 039 India
| |
Collapse
|
39
|
Kolarević A, Kocić G, Yancheva D, Šmelcerović A. IN SILICO PHARMACOKINETIC AND TOXICOLOGICAL STUDY OF DNASE INHIBITORS. ACTA MEDICA MEDIANAE 2016. [DOI: 10.5633/amm.2016.0401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
40
|
Jones LH, Heinis C. Chemical biology & drug discovery. Eur J Med Chem 2014; 88:1-2. [PMID: 25307206 DOI: 10.1016/j.ejmech.2014.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Lyn H Jones
- Worldwide Medicinal Chemistry, Pfizer BioTherapeutics Chemistry, Cambridge, MA, USA.
| | - Christian Heinis
- Laboratory of Therapeutic Proteins and Peptides (LPPT), École Polytechnique Fédérale de Lausanne (EPFL), BCH 5305 (Bâtochime), Lausanne CH-1015, Switzerland.
| |
Collapse
|
41
|
Kamdje AHN, Etet PFS, Vecchio L, Tagne RS, Amvene JM, Muller JM, Krampera M, Lukong KE. New targeted therapies for breast cancer: A focus on tumor microenvironmental signals and chemoresistant breast cancers. World J Clin Cases 2014; 2:769-786. [PMID: 25516852 PMCID: PMC4266825 DOI: 10.12998/wjcc.v2.i12.769] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/12/2014] [Accepted: 09/23/2014] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the most frequent female malignancy worldwide. Current strategies in breast cancer therapy, including classical chemotherapy, hormone therapy, and targeted therapies, are usually associated with chemoresistance and serious adverse effects. Advances in our understanding of changes affecting the interactome in advanced and chemoresistant breast tumors have provided novel therapeutic targets, including, cyclin dependent kinases, mammalian target of rapamycin, Notch, Wnt and Shh. Inhibitors of these molecules recently entered clinical trials in mono- and combination therapy in metastatic and chemo-resistant breast cancers. Anticancer epigenetic drugs, mainly histone deacetylase inhibitors and DNA methyltransferase inhibitors, also entered clinical trials. Because of the complexity and heterogeneity of breast cancer, the future in therapy lies in the application of individualized tailored regimens. Emerging therapeutic targets and the implications for personalized-based therapy development in breast cancer are herein discussed.
Collapse
|