1
|
Seinkmane E, Edmondson A, Peak-Chew SY, Zeng A, Rzechorzek NM, James NR, West J, Munns J, Wong DC, Beale AD, O'Neill JS. Circadian regulation of macromolecular complex turnover and proteome renewal. EMBO J 2024; 43:2813-2833. [PMID: 38778155 PMCID: PMC11217436 DOI: 10.1038/s44318-024-00121-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
Although costly to maintain, protein homeostasis is indispensable for normal cellular function and long-term health. In mammalian cells and tissues, daily variation in global protein synthesis has been observed, but its utility and consequences for proteome integrity are not fully understood. Using several different pulse-labelling strategies, here we gain direct insight into the relationship between protein synthesis and abundance proteome-wide. We show that protein degradation varies in-phase with protein synthesis, facilitating rhythms in turnover rather than abundance. This results in daily consolidation of proteome renewal whilst minimising changes in composition. Coupled rhythms in synthesis and turnover are especially salient to the assembly of macromolecular protein complexes, particularly the ribosome, the most abundant species of complex in the cell. Daily turnover and proteasomal degradation rhythms render cells and mice more sensitive to proteotoxic stress at specific times of day, potentially contributing to daily rhythms in the efficacy of proteasomal inhibitors against cancer. Our findings suggest that circadian rhythms function to minimise the bioenergetic cost of protein homeostasis through temporal consolidation of protein turnover.
Collapse
Affiliation(s)
- Estere Seinkmane
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Anna Edmondson
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Sew Y Peak-Chew
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Aiwei Zeng
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Nina M Rzechorzek
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Nathan R James
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - James West
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Jack Munns
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - David Cs Wong
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Andrew D Beale
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| | - John S O'Neill
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| |
Collapse
|
2
|
Vercammen H, Ondra M, Kotulova J, De La Hoz EC, Witters C, Jecmenova K, Le Compte M, Deben C, Ní Dhubhghaill S, Koppen C, Hajdúch M, Van den Bogerd B. "Keep on ROCKIn": Repurposed ROCK inhibitors to boost corneal endothelial regeneration. Biomed Pharmacother 2024; 174:116435. [PMID: 38513591 DOI: 10.1016/j.biopha.2024.116435] [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: 01/10/2024] [Revised: 03/06/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
The global shortage of corneal endothelial graft tissue necessitates the exploration of alternative therapeutic strategies. Rho-associated protein kinase inhibitors (ROCKi), recognized for their regenerative potential in cardiology, oncology, and neurology, have shown promise in corneal endothelial regeneration. This study investigates the repurposing potential of additional ROCKi compounds. Through screening a self-assembled library of ROCKi on B4G12 corneal endothelial cells, we evaluated their dose-dependent effects on proliferation, migration, and toxicity using live-cell imaging. Nine ROCKi candidates significantly enhanced B4G12 proliferation compared to the basal growth rate. These candidates were further assessed for their potential to accelerate wound closure as another indicator for tissue regeneration capacity, with most demonstrating notable efficacy. To assess the potential impact of candidate ROCKi on key corneal endothelial cell markers related to cell proliferation, leaky tight junctions and ion efflux capacity, we analyzed the protein expression of cyclin E1, CDK2, p16, ZO-1 and Na+/K+-ATPase, respectively. Immunocytochemistry and western blot analysis confirmed the preservation of corneal endothelial markers post-treatment with ROCKi hits. However, notable cytoplasm enlargement and nuclear fragmentation were detected after the treatment with SR-3677 and Thiazovivin, indicating possible cellular stress. In compared parameters, Chroman-1 at a concentration of 10 nM outperformed other ROCKi, requiring significantly 1000-fold lower effective concentration than established ROCKi Y-27632 and Fasudil. Altogether, this study underscores the potential of repurposing ROCKi for treating corneal endothelial dysfunctions, offering a viable alternative to conventional grafting methods, and highlights Chroman-1 as a promising candidate structure for hit-to-lead development.
Collapse
Affiliation(s)
- Hendrik Vercammen
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium; Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium; DrugVision Lab, University of Antwerp, Wilrijk, Belgium.
| | - Martin Ondra
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic; Czech Advanced Technology and Research Institute (CATRIN), Palacky University Olomouc, Olomouc, Czech Republic
| | - Jana Kotulova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | | | - Charissa Witters
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium; Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium; DrugVision Lab, University of Antwerp, Wilrijk, Belgium
| | - Katerina Jecmenova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | | | | | | | - Carina Koppen
- Antwerp Research Group for Ocular Science (ARGOS), Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium; Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic; Czech Advanced Technology and Research Institute (CATRIN), Palacky University Olomouc, Olomouc, Czech Republic
| | - Bert Van den Bogerd
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic; Czech Advanced Technology and Research Institute (CATRIN), Palacky University Olomouc, Olomouc, Czech Republic.
| |
Collapse
|
3
|
Ortmann W, Such A, Kolaczkowska E. Impact of microparticles released during murine systemic inflammation on macrophage activity and reactive nitrogen species regulation. Immunol Res 2024; 72:299-319. [PMID: 38008825 PMCID: PMC11031483 DOI: 10.1007/s12026-023-09436-7] [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: 04/06/2023] [Accepted: 11/13/2023] [Indexed: 11/28/2023]
Abstract
Microparticles (MPs) packaged with numerous bioactive molecules are essential vehicles in cellular communication in various pathological conditions, including systemic inflammation, Whereas MPs are studied mostly upon isolation, their detection in vivo is limited. Impact of MPs might depend on target cell type and cargo they carry; thus herein, we aimed at verifying MPs' impact on macrophages. Unlike neutrophils, monocytes/macrophages are rather inactive during sepsis, and we hypothesized this might be at least partially controlled by MPs. For the above reasons, we focused on the detection of MPs with intravital microscopy (IVM) and report the presence of putative neutrophil-derived MPs in the vasculature of cremaster muscle of endotoxemic mice. Subsequently, we characterized MPs isolated not only from their blood but also from the peritoneal cavity and observed differences in their size, concentration, and cargo. Such MPs were then used to study their impact on RAW 264.7 macrophage cell line performance (cell viability/activity, cytokines, oxygen, and nitrogen reactive species). Addition of MPs to macrophages with or without co-stimulation with lipopolysaccharide did not affect respiratory burst, somewhat decreased mitochondrial activity but increased inducible nitric oxide synthase (iNOS) expression, and NO production especially in case of plasma-derived MPs. The latter MPs carried more iNOS-controlling ceruloplasmin than those discharged into the peritoneal cavity. We conclude that MPs can be detected in vivo with IVM and their cellular origin identified. They are heterogeneous in nature depending on the site of their release. Consequently, microparticles released during systemic inflammation to various body compartments differentially affect macrophages.
Collapse
Affiliation(s)
- Weronika Ortmann
- Laboratory of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9 Street, 30-387, Krakow, Poland
| | - Anna Such
- Laboratory of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9 Street, 30-387, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Elzbieta Kolaczkowska
- Laboratory of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9 Street, 30-387, Krakow, Poland.
| |
Collapse
|
4
|
Alfatah M, Zhang Y, Naaz A, Cheng TYN, Eisenhaber F. PICLS with human cells is the first high throughput screening method for identifying novel compounds that extend lifespan. Biol Direct 2024; 19:8. [PMID: 38254217 PMCID: PMC10804585 DOI: 10.1186/s13062-024-00455-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/18/2024] [Indexed: 01/24/2024] Open
Abstract
Gerontology research on anti-aging interventions with drugs could be an answer to age-related diseases, aiming at closing the gap between lifespan and healthspan. Here, we present two methods for assaying chronological lifespan in human cells: (1) a version of the classical outgrowth assay with quantitative assessment of surviving cells and (2) a version of the PICLS method (propidium iodide fluorescent-based measurement of cell death). Both methods are fast, simple to conduct, cost-effective, produce quantitative data for further analysis and can be used with diverse human cell lines. Whereas the first method is ideal for validation and testing the post-intervention reproductive potential of surviving cells, the second method has true high-throughput screening potential. The new technologies were validated with known anti-aging compounds (2,5-anhydro-D-mannitol and rapamycin). Using the high-throughput screening method, we screened a library of 162 chemical entities and identified three compounds that extend the longevity of human cells.
Collapse
Affiliation(s)
- Mohammad Alfatah
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix #07-01, Singapore, 138671, Republic of Singapore.
| | - Yizhong Zhang
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix #07-01, Singapore, 138671, Republic of Singapore
| | - Arshia Naaz
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome #02-01, Singapore, 138672, Republic of Singapore
| | - Trishia Yi Ning Cheng
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix #07-01, Singapore, 138671, Republic of Singapore
| | - Frank Eisenhaber
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix #07-01, Singapore, 138671, Republic of Singapore
- LASA - Lausitz Advanced Scientific Applications gGmbH, Straße der Einheit 2-24, 02943, Weißwasser, Federal Republic of Germany
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), Singapore, 637551, Republic of Singapore
| |
Collapse
|
5
|
Nawrot-Hadzik I, Matkowski A, Fast M, Choromańska A. The combination of pro-oxidative acting vanicosides and GLUT1 inhibitor (WZB117) exerts a synergistic cytotoxic effect against melanoma cells. Fitoterapia 2023; 171:105702. [PMID: 37848084 DOI: 10.1016/j.fitote.2023.105702] [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: 05/09/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/19/2023]
Abstract
Vanicosides A and B isolated from Reynoutria sachalinensis rhizomes are disaccharide phenylpropanoid esters with proven antioxidant activity. Our earlier study showed the cytotoxic activity of vanicosides against melanoma cells, but the mechanism of cell death has not been elucidated. Based on the chemical structure of vanicosides, we proposed that they may induce cell death by generating reactive oxygen species (ROS) into melanoma cells. Moreover, the glucose molecule in their structure can affect the glucose transporters (GLUTs), upregulated in cancer cells. The A375 (melanotic) and C32 (amelanotic) melanoma cell lines were applied. Cell viability assay and ROS-Glo™ assay were performed before and after blocking of Glucose Transporter Type 1 (GLUT1) by WZB117. Fibroblasts and the SKOV-3 line were included in the study to test selectivity in the action of vanicosides and help to elucidate the mechanism of action. Upon incubation with vanicosides, high production of ROS occured, especially inside C32 cells, which was significantly reduced after GLUT-1 blocking. The A375 cells produced less ROS. Melanoma cells were simillary sensitive to the cytotoxic effects of vanicosides, which was clearly enhanced when vanicosides were used together with the WZB117 (GLUT1 inhibitor). The SKOV-3 line and the fibroblasts showed much less sensitivity to the cytotoxicity of vanicosides, also used together with WZB117. Moreover, no significant ROS formation was observed in these lines. The study proved that vanicosides generate ROS inside melanoma cells. These findings suggest that the combination of pro-oxidative acting vanicosides and GLUT1 inhibitors exerts a synergistic cytotoxic effect on melanoma cells.
Collapse
Affiliation(s)
- Izabela Nawrot-Hadzik
- Department of Pharmaceutical Biology and Biotechnology, Division of Pharmaceutical Biology and Botany, Wroclaw Medical University, 50-556 Wroclaw, Poland.
| | - Adam Matkowski
- Department of Pharmaceutical Biology and Biotechnology, Division of Pharmaceutical Biology and Botany, Wroclaw Medical University, 50-556 Wroclaw, Poland.
| | - Magdalena Fast
- Department of Pharmaceutical Biology and Biotechnology, Division of Pharmaceutical Biology and Botany, Wroclaw Medical University, 50-556 Wroclaw, Poland.
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland.
| |
Collapse
|
6
|
Dergilev K, Zubkova E, Guseva A, Tsokolaeva Z, Goltseva Y, Beloglazova I, Ratner E, Andreev A, Partigulov S, Lepilin M, Menshikov M, Parfyonova Y. Tumor Necrosis Factor-Alpha Induces Proangiogenic Profiling of Cardiosphere-Derived Cell Secretome and Increases Its Ability to Stimulate Angiogenic Properties of Endothelial Cells. Int J Mol Sci 2023; 24:16575. [PMID: 38068898 PMCID: PMC10706276 DOI: 10.3390/ijms242316575] [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: 09/29/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Ischemic heart disease and its complications, such as myocardial infarction and heart failure, are the leading causes of death in modern society. The adult heart innately lacks the capacity to regenerate the damaged myocardium after ischemic injury. Multiple lines of evidence indicated that stem-cell-based transplantation is one of the most promising treatments for damaged myocardial tissue. Different kinds of stem cells have their advantages for treating ischemic heart disease. One facet of their mechanism is the paracrine effect of the transplanted cells. Particularly promising are stem cells derived from cardiac tissue per se, referred to as cardiosphere-derived cells (CDCs), whose therapeutic effect is mediated by the paracrine mechanism through secretion of multiple bioactive molecules providing immunomodulatory, angiogenic, anti-fibrotic, and anti-inflammatory effects. Although secretome-based therapies are increasingly being used to treat various cardiac pathologies, many obstacles remain because of population heterogeneity, insufficient understanding of potential modulating compounds, and the principles of secretome regulation, which greatly limit the feasibility of this technology. In addition, components of the inflammatory microenvironment in ischemic myocardium may influence the secretome content of transplanted CDCs, thus altering the efficacy of cell therapy. In this work, we studied how Tumor necrosis factor alpha (TNFa), as a key component of the pro-inflammatory microenvironment in damaged myocardium from ischemic injury and heart failure, may affect the secretome content of CDCs and their angiogenic properties. We have shown for the first time that TNFa may act as a promising compound modulating the CDC secretome, which induces its profiling to enhance proangiogenic effects on endothelial cells. These results allow us to elucidate the underlying mechanisms of the impact of the inflammatory microenvironment on transplanted CDCs and may contribute to the optimization of CDC efficiency and the development of the technology for producing the CDC secretome with enhanced proangiogenic properties for cell-free therapy.
Collapse
Affiliation(s)
- Konstantin Dergilev
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Ekaterina Zubkova
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Alika Guseva
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Zoya Tsokolaeva
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 141534 Moscow, Russia
| | - Yulia Goltseva
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Irina Beloglazova
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Elizaveta Ratner
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Alexander Andreev
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Stanislav Partigulov
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Mikhail Lepilin
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Mikhail Menshikov
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Yelena Parfyonova
- Federal State Budgetary, Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| |
Collapse
|
7
|
Kardorff M, Mahler HC, Huwyler J, Sorret L. Comparison of cell viability methods for human mesenchymal/stromal stem cells and human A549 lung carcinoma cells after freeze-thaw stress. J Pharmacol Toxicol Methods 2023; 124:107474. [PMID: 37866798 DOI: 10.1016/j.vascn.2023.107474] [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: 06/25/2023] [Revised: 09/27/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
For the safety and efficacy of frozen cell therapy products, determination of cellular viability is key. However, results of cell viability measurements do not only depend on the cell line or on the inflicted stress, but also on the assay used, making inter-experimental comparisons difficult. The aim of this study was thus to assess commonly used viability assays in clinically relevant human mesenchymal/stromal stem cells and human A549 lung carcinoma cells. Post freeze-thaw stress viability and proliferation were evaluated under different conditions using trypan blue, acridine orange/DAPI stain, alamarBlue, ATP, and neutral red assays. Significant differences in cell viability between metabolic assays were observed, likely due to their distinct intrinsic detection mechanisms. Membrane-integrity based assays generally overestimated cell viabilities in this study. Furthermore, noticeable differences in inter-assay sensitivities were observed. These differences highlight that cell viability methods should be meticulously selected and their associated results carefully interpreted in a relevant context to ensure reliable conclusions. Indeed, although cell membrane integrity based assays are a popular choice to determine cellular quality attributes after freezing and thawing, we demonstrate that metabolic assays may be more suitable in this context.
Collapse
Affiliation(s)
- Markus Kardorff
- Drug Product Services, Lonza AG, Hochbergerstrasse 60G, 4057 Basel, Switzerland; Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | | | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Léa Sorret
- Drug Product Services, Lonza AG, Hochbergerstrasse 60G, 4057 Basel, Switzerland.
| |
Collapse
|
8
|
Abdel Rahman DE, Fouad MA, Mohammed ER, El-Zoheiry HH, Abdelrasheed Allam H. Novel VEGFR-2 inhibitors as antiangiogenic and apoptotic agents via paracrine and autocrine cascades: Design, synthesis, and biological evaluation. Bioorg Chem 2023; 139:106678. [PMID: 37354661 DOI: 10.1016/j.bioorg.2023.106678] [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: 04/18/2023] [Revised: 05/31/2023] [Accepted: 06/12/2023] [Indexed: 06/26/2023]
Abstract
Appertaining to its paracrine and autocrine signaling loops, VEGFR-2 succeeded in grabbing attention as one of the leading targets in cancer treatment. Based on the foregoing and our comprehensive studies regarding pharmacophoric features and activity of sorafenib, novel phenylpyridazinone based VEGFR-2 inhibitors 4, 6a-e, 7a,b, 9a,b, 12a-c, 13a,b, 14a,b, 15a,b, and 17a-d were optimized. An assortment of biological assays was conducted to assess the antiangiogenic and apoptotic activities of the synthesized derivatives. In vitro VEGFR-2 kinase assay verified the inhibitory activity of the synthesized derivatives with IC50 values from 49.1 to 418.0 nM relative to the reference drug sorafenib (IC50 = 81.8 nM). Antiproliferative activity against HUVECs revealed that compounds 2-{2-[2-(6-oxo-3-phenylpyridazin-1(6H)-yl)acetyl]hydrazineyl}-N-(p-tolyl)acetamide (12c) and 2-[(5-mercapto-4-methyl-4H-1,2,4-triazol-3-yl)methyl]-6-phenylpyridazin-3(2H)-one (13a) possessed superior activity (IC50 values = 11.5 and 12.3 nM, respectively) in comparison to sorafenib (IC50 = 23.2 nM). For the purpose of appraising their antiproliferative effect, derivatives 12c and 13a were exposed to cell cycle analysis, apoptotic, cell invasion and migration assays in addition to determination of VEGFR-2 in protein level. Moreover, cytotoxicity as well as selectivity index against WI-38 cell line was measured to examine safety of derivatives 12c and 13a. After that, molecular docking study was executed on the top five compounds in the in vitro VEGFR-2 kinase assay 6d, 12c, 13a, 14a and 17c to get a deep perception on binding mode of the synthesized compounds and correlate the design strategy with biological results. Finally, physicochemical, pharmacokinetic properties, and drug-likeness studies were performed on the top five derivative in in vitro VEGFR-2 kinase assay.
Collapse
Affiliation(s)
- Doaa E Abdel Rahman
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| | - Marwa A Fouad
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt; Pharmaceutical Chemistry Department, School of Pharmacy, New Giza University, Newgiza, km 22 Cairo-Alexandria Desert Road, Cairo, Egypt
| | - Eman R Mohammed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| | - Haidy H El-Zoheiry
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt.
| | - Heba Abdelrasheed Allam
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| |
Collapse
|
9
|
Kim S, Park J, Ho JN, Kim D, Lee S, Jeon JS. 3D vascularized microphysiological system for investigation of tumor-endothelial crosstalk in anti-cancer drug resistance. Biofabrication 2023; 15:045016. [PMID: 37567223 DOI: 10.1088/1758-5090/acef99] [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: 04/26/2023] [Accepted: 08/11/2023] [Indexed: 08/13/2023]
Abstract
Despite the advantages of microfluidic system in drug screening, vascular systems responsible for the transport of drugs and nutrients have been hardly considered in the microfluidic-based chemotherapeutic screening. Considering the physiological characteristics of highly vascularized urinary tumors, we here investigated the chemotherapeutic response of bladder tumor cells using a vascularized tumor on a chip. The microfluidic chip was designed to have open-top region for tumor sample introduction and hydrophilic rail for spontaneous hydrogel patterning, which contributed to the construction of tumor-hydrogel-endothelium interfaces in a spatiotemporal on-demand manner. Utilizing the chip where intravascularly injected cisplatin diffuse across the endothelium and transport into tumor samples, chemotherapeutic responses of cisplatin-resistant or -susceptible bladder tumor cells were evaluated, showing the preservation of cellular drug resistance even within the chip. The open-top structure also enabled the direct harvest of tumor samples and post analysis in terms of secretome and gene expressions. Comparing the cisplatin efficacy of the cisplatin-resistant tumor cells in the presence or absence of endothelium, we found that the proliferation rates of tumor cells were increased in the vasculature-incorporated chip. These have suggested that our vascularized tumor chip allows the establishment of vascular-gel-tumor interfaces in spatiotemporal manners and further enables investigations of chemotherapeutic screening.
Collapse
Affiliation(s)
- Seunggyu Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States of America
| | - Joonha Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jin-Nyoung Ho
- Department of Urology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
| | - Danhyo Kim
- Department of Urology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
| | - Sangchul Lee
- Department of Urology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
| | - Jessie S Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
10
|
Lie KCM, Bonturi CR, Salu BR, de Oliveira JR, Bonini Galo M, Paiva PMG, Correia MTDS, Oliva MLV. Impairment of SK-MEL-28 Development-A Human Melanoma Cell Line-By the Crataeva tapia Bark Lectin and Its Sequence-Derived Peptides. Int J Mol Sci 2023; 24:10617. [PMID: 37445794 DOI: 10.3390/ijms241310617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Melanoma is difficult to treat with chemotherapy, prompting the need for new treatments. Protease inhibitors have emerged as promising candidates as tumor cell proteases promote metastasis. Researchers have developed a chimeric form of the Bauhinia bauhinioides kallikrein inhibitor, rBbKIm, which has shown negative effects on prostate tumor cell lines DU145 and PC3. Crataeva tapia bark lectin, CrataBL, targets sulfated oligosaccharides in glycosylated proteins and has also demonstrated deleterious effects on prostate and glioblastoma tumor cells. However, neither rBbKIm nor its derived peptides affected the viability of SK-MEL-28, a melanoma cell line, while CrataBL decreased viability by over 60%. Two peptides, Pep. 26 (Ac-Q-N-S-S-L-K-V-V-P-L-NH2) and Pep. 27 (Ac-L-P-V-V-K-L-S-S-N-Q-NH2), were also tested. Pep. 27 suppressed cell migration and induced apoptosis when combined with vemurafenib, while Pep. 26 inhibited cell migration and reduced nitric oxide and the number of viable cells. Vemurafenib, a chemotherapy drug used to treat melanoma, was found to decrease the release of interleukin 8 and PDGF-AB/BB cytokines and potentiated the effects of proteins and peptides in reducing these cytokines. These findings suggest that protease inhibitors may be effective in blocking melanoma cells and highlight the potential of CrataBL and its derived peptides.
Collapse
Affiliation(s)
| | - Camila Ramalho Bonturi
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | - Bruno Ramos Salu
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | | | - Márcia Bonini Galo
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | | | | | | |
Collapse
|
11
|
Kohl Y, William N, Elje E, Backes N, Rothbauer M, Srancikova A, Rundén-Pran E, El Yamani N, Korenstein R, Madi L, Barbul A, Kozics K, Sramkova M, Steenson K, Gabelova A, Ertl P, Dusinska M, Nelson A. Rapid identification of in vitro cell toxicity using an electrochemical membrane screening platform. Bioelectrochemistry 2023; 153:108467. [PMID: 37244203 DOI: 10.1016/j.bioelechem.2023.108467] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/28/2023] [Accepted: 05/15/2023] [Indexed: 05/29/2023]
Abstract
This study compares the performance and output of an electrochemical phospholipid membrane platform against respective in vitro cell-based toxicity testing methods using three toxicants of different biological action (chlorpromazine (CPZ), colchicine (COL) and methyl methanesulphonate (MMS)). Human cell lines from seven different tissues (lung, liver, kidney, placenta, intestine, immune system) were used to validate this physicochemical testing system. For the cell-based systems, the effective concentration at 50 % cell death (EC50) values are calculated. For the membrane sensor, a limit of detection (LoD) value was extracted as a quantitative parameter describing the minimum concentration of toxicant which significantly affects the structure of the phospholipid sensor membrane layer. LoD values were found to align well with the EC50 values when acute cell viability was used as an end-point and showed a similar toxicity ranking of the tested toxicants. Using the colony forming efficiency (CFE) or DNA damage as end-point, a different order of toxicity ranking was observed. The results of this study showed that the electrochemical membrane sensor generates a parameter relating to biomembrane damage, which is the predominant factor in decreasing cell viability when in vitro models are acutely exposed to toxicants. These results lead the way to using electrochemical membrane-based sensors for rapid relevant preliminary toxicity screens.
Collapse
Affiliation(s)
- Yvonne Kohl
- Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, Sulzbach 66280, Germany.
| | - Nicola William
- School of Chemistry and Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom.
| | - Elisabeth Elje
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller 2007, Norway; Faculty of Medicine, Institute of Basic Medical Sciences Department of Molecular Medicine, University of Oslo, Sognsvannsveien 9, Oslo 0372, Norway.
| | - Nadine Backes
- Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, Sulzbach 66280, Germany
| | - Mario Rothbauer
- Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Annamaria Srancikova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| | - Elise Rundén-Pran
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller 2007, Norway.
| | - Naouale El Yamani
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller 2007, Norway
| | - Rafi Korenstein
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Lea Madi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Alexander Barbul
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Katarina Kozics
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| | - Monika Sramkova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| | - Karen Steenson
- School of Chemistry and Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom.
| | - Alena Gabelova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| | - Peter Ertl
- Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria; Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Maria Dusinska
- NILU-Norwegian Institute for Air Research, Department for Environmental Chemistry, Health Effects Laboratory, Instituttveien 18, Kjeller 2007, Norway.
| | - Andrew Nelson
- School of Chemistry and Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom.
| |
Collapse
|
12
|
Sremački I, Asadian M, De Geyter N, Leys C, Geris L, Nikiforov A. Potentials of a Plasma-Aerosol System for Wound Healing Advanced by Drug Introduction: An In Vitro Study. ACS Biomater Sci Eng 2023; 9:2392-2407. [PMID: 37129346 DOI: 10.1021/acsbiomaterials.2c01391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Cold plasmas have found their application in a wide range of biomedical fields by virtue of their high chemical reactivity. In the past decades, many attempts have been made to use cold plasmas in wound healing, and within this field, many studies have focused on plasma-induced cell proliferation mechanisms. In this work, one step further has been taken to demonstrate the advanced role of plasma in wound healing. To this end, the simultaneous ability of plasma to induce cell proliferation and permeabilize treated cells has been examined in the current study. The driving force was to advance the wound healing effect of plasma with drug delivery. On this subject, we demonstrate in vitro the healing effect of Ar, Ar+N2 plasma, and their aerosol counterparts. A systematic study has been carried out to study the role of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in cell adhesion, signaling, differentiation, and proliferation. An additional investigation was also performed to study the permeabilization of cells and the delivery of the modeled drug carrier fluorescein isothiocyanate (FITC) labeled dextran into cells upon plasma treatment. Short 35 s plasma treatments were found to promote fibroblast adhesion, migration, signaling, proliferation, and differentiation by means of reactive oxygen and nitrogen species (RONS) created by plasma and deposited into the cell environment. The impact of the plasma downstream products NO2- and NO3- on the expressions of the focal adhesion's genes, syndecans, and collagens was observed to be prominent. On the other hand, the differentiation of fibroblasts to myofibroblasts was mainly initiated by ROS produced by the plasma. In addition, the ability of plasma to locally permeabilize fibroblast cells was demonstrated. During proliferative cell treatment, plasma can simultaneously induce cell membrane permeabilization (d ∼ 7.3 nm) by the species OH and H2O2. The choice for a plasma or a plasma-aerosol configuration thus allows the possibility to change the spatial chemistry of drug delivery molecules and thus to locally deliver drugs. Accordingly, this study offers a pivotal step toward plasma-assisted wound healing advanced by drug delivery.
Collapse
Affiliation(s)
- Ivana Sremački
- Department of Applied Physics, Ghent University, Sint-Pietersnieuwstraat 41, Gent 9000, Belgium
| | - Mahtab Asadian
- Department of Applied Physics, Ghent University, Sint-Pietersnieuwstraat 41, Gent 9000, Belgium
- Skeletal Biology & Engineering Research Center, ON1 Herestraat 49, 3000 Leuven, Belgium
| | - Nathalie De Geyter
- Department of Applied Physics, Ghent University, Sint-Pietersnieuwstraat 41, Gent 9000, Belgium
| | - Christophe Leys
- Department of Applied Physics, Ghent University, Sint-Pietersnieuwstraat 41, Gent 9000, Belgium
| | - Liesbet Geris
- Skeletal Biology & Engineering Research Center, ON1 Herestraat 49, 3000 Leuven, Belgium
- Biomechanics Research Unit, Liège University, GIGA In Silico Medicine, Quartier Hôpital avenue de l'Hôpital 11, 4000 Liège, Belgium
| | - Anton Nikiforov
- Department of Applied Physics, Ghent University, Sint-Pietersnieuwstraat 41, Gent 9000, Belgium
| |
Collapse
|
13
|
Kobori T, Ito Y, Sawada Y, Urashima Y, Ito T, Obata T. Cellular Membrane Localization of Innate Immune Checkpoint Molecule CD47 Is Regulated by Radixin in Human Pancreatic Ductal Adenocarcinoma Cells. Biomedicines 2023; 11:biomedicines11041117. [PMID: 37189735 DOI: 10.3390/biomedicines11041117] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/29/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
In the past decade, immune checkpoint inhibitors have exhibited potent antitumor efficacy against multiple solid malignancies but limited efficacy against pancreatic ductal adenocarcinoma (PDAC). Cluster of differentiation (CD) 47, a member of the immunoglobulin G superfamily, is overexpressed in the surface membrane of PDAC and independently correlates with a worse clinical prognosis. Furthermore, CD47 functions as a dominant macrophage checkpoint, providing a potent "do not eat me" signal to enable cancer cells to evade the innate immune system. Thus, the blockade of CD47 is a promising immunotherapeutic strategy for PDAC. In this study, we determined whether ezrin/radixin/moesin (ERM) family members, which post-translationally modulate the cellular membrane localization of numerous transmembrane proteins by crosslinking with the actin cytoskeleton, contribute to the cellular membrane localization of CD47 in KP-2 cells derived from human PDAC. Immunofluorescence analysis showed that CD47 and ezrin/radixin were highly co-localized in the plasma membrane. Interestingly, gene silencing of radixin but not ezrin dramatically decreased the cell surface expression of CD47 but had little effects on its mRNA level. Furthermore, CD47 and radixin interacted with each other, as determined by a co-immunoprecipitation assay. In conclusion, radixin regulates the cellular membrane localization of CD47 as a scaffold protein in KP-2 cells.
Collapse
Affiliation(s)
- Takuro Kobori
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Osaka 584-8540, Japan
| | - Yui Ito
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Osaka 584-8540, Japan
| | - Yuka Sawada
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Osaka 584-8540, Japan
| | - Yoko Urashima
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Osaka 584-8540, Japan
| | - Takuya Ito
- Laboratory of Natural Medicines, Faculty of Pharmacy, Osaka Ohtani University, Osaka 584-8540, Japan
| | - Tokio Obata
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Osaka 584-8540, Japan
| |
Collapse
|
14
|
Effect of Electrospun PLGA/Collagen Scaffolds on Cell Adhesion, Viability, and Collagen Release: Potential Applications in Tissue Engineering. Polymers (Basel) 2023; 15:polym15051079. [PMID: 36904322 PMCID: PMC10006987 DOI: 10.3390/polym15051079] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
The development of scaffolding obtained by electrospinning is widely used in tissue engineering due to porous and fibrous structures that can mimic the extracellular matrix. In this study, poly (lactic-co-glycolic acid) (PLGA)/collagen fibers were fabricated by electrospinning method and then evaluated in the cell adhesion and viability of human cervical carcinoma HeLa and NIH-3T3 fibroblast for potential application in tissue regeneration. Additionally, collagen release was assessed in NIH-3T3 fibroblasts. The fibrillar morphology of PLGA/collagen fibers was verified by scanning electron microscopy. The fiber diameter decreased in the fibers (PLGA/collagen) up to 0.6 µm. FT-IR spectroscopy and thermal analysis confirmed that both the electrospinning process and the blend with PLGA give structural stability to collagen. Incorporating collagen in the PLGA matrix promotes an increase in the material's rigidity, showing an increase in the elastic modulus (38%) and tensile strength (70%) compared to pure PLGA. PLGA and PLGA/collagen fibers were found to provide a suitable environment for the adhesion and growth of HeLa and NIH-3T3 cell lines as well as stimulate collagen release. We conclude that these scaffolds could be very effective as biocompatible materials for extracellular matrix regeneration, suggesting their potential applications in tissue bioengineering.
Collapse
|
15
|
Yamaura K, Sather NA, Metlushko A, Nishimura H, Pavlović RZ, Hambright S, Ravuri SK, Philippon MJ, Stupp SI, Bahney CS, Huard J. Sustained-release losartan from peptide nanofibers promotes chondrogenesis. Front Bioeng Biotechnol 2023; 11:1122456. [PMID: 36814717 PMCID: PMC9939695 DOI: 10.3389/fbioe.2023.1122456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/20/2023] [Indexed: 02/09/2023] Open
Abstract
Introduction: The central pathologic feature of osteoarthritis (OA) is the progressive loss of articular cartilage, which has a limited regenerative capacity. The TGF-β1 inhibitor, losartan, can improve cartilage repair by promoting hyaline rather that fibrous cartilage tissue regeneration. However, there are concerns about side effects associated with oral administration and short retention within the joint following intra-articular injections. To facilitate local and sustained intra-articular losartan delivery we have designed an injectable peptide amphiphile (PA) nanofiber that binds losartan. The aims of this study are to characterize the release kinetics of losartan from two different PA nanofiber compositions followed by testing pro-regenerative bioactivity on chondrocytes. Methods: We tested the impact of electrostatic interactions on nanostructure morphology and release kinetics of the negatively charged losartan molecule from either a positively or negatively charged PA nanofiber. Subsequently, cytotoxicity and bioactivity were evaluated in vitro in both normal and an IL-1β-induced OA chondrocyte model using ATDC5. Results: Both nanofiber systems promoted cell proliferation but that the positively-charged nanofibers also significantly increased glycosaminoglycans production. Furthermore, gene expression analysis suggested that losartan-encapsulated nanofibers had significant anti-inflammatory, anti-degenerative, and cartilage regenerative effects by significantly blocking TGF-β1 in this in vitro system. Discussion: The results of this study demonstrated that positively charged losartan sustained-release nanofibers may be a novel and useful treatment for cartilage regeneration and OA by blocking TGF-β1.
Collapse
Affiliation(s)
- Kohei Yamaura
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, United States
| | - Nicholas A. Sather
- Simpson Querrey Institute for Bionanotechnology, Northwestern University, Chicago, IL, United States
| | - Anna Metlushko
- Simpson Querrey Institute for Bionanotechnology, Northwestern University, Chicago, IL, United States
| | - Haruki Nishimura
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, United States
| | - Radoslav Z. Pavlović
- Simpson Querrey Institute for Bionanotechnology, Northwestern University, Chicago, IL, United States
| | - Sealy Hambright
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, United States
| | - Sudheer K. Ravuri
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, United States
| | - Marc J. Philippon
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, United States,The Steadman Clinic, Vail, CO, United States
| | - Samuel I. Stupp
- Simpson Querrey Institute for Bionanotechnology, Northwestern University, Chicago, IL, United States
| | - Chelsea S. Bahney
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, United States,The Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States,*Correspondence: Chelsea S. Bahney, ; Johnny Huard,
| | - Johnny Huard
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, United States,*Correspondence: Chelsea S. Bahney, ; Johnny Huard,
| |
Collapse
|
16
|
Cordeiro R, Alvites RD, Sousa AC, Lopes B, Sousa P, Maurício AC, Alves N, Moura C. Cellulose-Based Scaffolds: A Comparative Study for Potential Application in Articular Cartilage. Polymers (Basel) 2023; 15:781. [PMID: 36772083 PMCID: PMC9919712 DOI: 10.3390/polym15030781] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Osteoarthritis is a highly prevalent disease worldwide that leads to cartilage loss. Tissue engineering, involving scaffolds, cells, and stimuli, has shown to be a promising strategy for its repair. Thus, this study aims to manufacture and characterise different scaffolds with poly(ε-caprolactone) (PCL) with commercial cellulose (microcrystalline (McC) and methyl cellulose (MC) or cellulose from agro-industrial residues (corncob (CcC)) and at different percentages, 1%, 2%, and 3%. PCL scaffolds were used as a control. Morphologically, the produced scaffolds presented porosities within the desired for cell incorporation (57% to 65%). When submitted to mechanical tests, the incorporation of cellulose affects the compression resistance of the majority of scaffolds. Regarding tensile strength, McC2% showed the highest values. It was proven that all manufactured scaffolds suffered degradation after 7 days of testing because of enzymatic reactions. This degradation may be due to the dissolution of PCL in the organic solvent. Biological tests revealed that PCL, CcC1%, and McC3% are the best materials to combine with human dental pulp stem/stromal cells. Overall, results suggest that cellulose incorporation in PCL scaffolds promotes cellular adhesion/proliferation. Methyl cellulose scaffolds demonstrated some advantageous compressive properties (closer to native cartilaginous tissue) to proceed to further studies for application in cartilage repair.
Collapse
Affiliation(s)
- Rachel Cordeiro
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, 2430-028 Marinha Grande, Portugal
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
| | - Rui D. Alvites
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
- Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), University of Porto, Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Ana C. Sousa
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
- Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), University of Porto, Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Bruna Lopes
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
- Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), University of Porto, Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Patrícia Sousa
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
- Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), University of Porto, Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Ana C. Maurício
- Veterinary Clinics Department, Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
- Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), University of Porto, Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Nuno Alves
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, 2430-028 Marinha Grande, Portugal
- Associate Laboratory for Advanced Production and Intelligent Systems (ARISE), 4050-313 Porto, Portugal
| | - Carla Moura
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, 2430-028 Marinha Grande, Portugal
- Associate Laboratory for Advanced Production and Intelligent Systems (ARISE), 4050-313 Porto, Portugal
- Applied Research Institute (i2A), Polytechnic Institute of Coimbra, Rua da Misericórdia, Lagar dos Cortiços–S. Martinho do Bispo, 3045-093 Coimbra, Portugal
| |
Collapse
|
17
|
Wang R, Huang X, Zoetebier B, Dijkstra PJ, Karperien M. Enzymatic co-crosslinking of star-shaped poly(ethylene glycol) tyramine and hyaluronic acid tyramine conjugates provides elastic biocompatible and biodegradable hydrogels. Bioact Mater 2023; 20:53-63. [PMID: 35633871 PMCID: PMC9127275 DOI: 10.1016/j.bioactmat.2022.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/02/2022] Open
Abstract
A combination of the viscoelastic properties of hyaluronic acid (HA) and the elastic properties of star shaped 8-arm poly(ethylene glycol) (8-arm PEG) was used to design in-situ forming hydrogels. Hydrogels were prepared by the enzymatic crosslinking of a partially tyramine modified 8-arm PEG and a tyramine conjugated HA using horseradish peroxidase in the presence of hydrogen peroxide. Hydrogels of the homopolymer conjugates and mixtures thereof were rapidly formed within seconds under physiological conditions at low polymer and enzyme concentrations. Elastic hydrogels with high gel content (≥95%) and high storage moduli (up to 22.4 kPa) were obtained. An in vitro study in the presence of hyaluronidase (100 U/mL) revealed that with increasing PEG content the degradation time of the hybrid hydrogels increased up to several weeks, whereas hydrogels composed of only hyaluronic acid degraded within 2 weeks. Human mesenchymal stem cells (hMSCs) incorporated in the hybrid hydrogels remained viable as shown by a PrestoBlue and a live-dead assay, confirming the biocompatibility of the constructs. The production of an extracellular matrix by re-differentiation of encapsulated human chondrocytes was followed over a period of 28 days. Gene expression indicated that these highly elastic hydrogels induced an enhanced production of collagen type II. At low PEG-TA/HA-TA ratios a higher expression of SOX 9 and ACAN was observed. These results indicate that by modulating the ratio of PEG/HA, injectable hydrogels can be prepared applicable as scaffolds for tissue regeneration applications.
Collapse
Affiliation(s)
| | | | - Bram Zoetebier
- Department of Developmental BioEngineering, Faculty of Science and Technology, Tech Med Centre, University of Twente, P.O. Box 217, 7500 AE, Enschede, the Netherlands
| | - Pieter J. Dijkstra
- Department of Developmental BioEngineering, Faculty of Science and Technology, Tech Med Centre, University of Twente, P.O. Box 217, 7500 AE, Enschede, the Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, Faculty of Science and Technology, Tech Med Centre, University of Twente, P.O. Box 217, 7500 AE, Enschede, the Netherlands
| |
Collapse
|
18
|
Highlights of New Strategies to Increase the Efficacy of Transition Metal Complexes for Cancer Treatments. Molecules 2022; 28:molecules28010273. [PMID: 36615466 PMCID: PMC9822110 DOI: 10.3390/molecules28010273] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023] Open
Abstract
Although important progress has been made, cancer still remains a complex disease to treat. Serious side effects, the insurgence of resistance and poor selectivity are some of the problems associated with the classical metal-based anti-cancer therapies currently in clinical use. New treatment approaches are still needed to increase cancer patient survival without cancer recurrence. Herein, we reviewed two promising-at least in our opinion-new strategies to increase the efficacy of transition metal-based complexes. First, we considered the possibility of assembling two biologically active fragments containing different metal centres into the same molecule, thus obtaining a heterobimetallic complex. A critical comparison with the monometallic counterparts was done. The reviewed literature has been divided into two groups: the case of platinum; the case of gold. Secondly, the conjugation of metal-based complexes to a targeting moiety was discussed. Particularly, we highlighted some interesting examples of compounds targeting cancer cell organelles according to a third-order targeting approach, and complexes targeting the whole cancer cell, according to a second-order targeting strategy.
Collapse
|
19
|
Ashraf J, Lau S, Akbarinejad A, Evans CW, Williams DE, Barker D, Travas-Sejdic J. Conducting Polymer-Infused Electrospun Fibre Mat Modified by POEGMA Brushes as Antifouling Biointerface. BIOSENSORS 2022; 12:1143. [PMID: 36551110 PMCID: PMC9775683 DOI: 10.3390/bios12121143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Biofouling on surfaces, caused by the assimilation of proteins, peptides, lipids and microorganisms, leads to contamination, deterioration and failure of biomedical devices and causes implants rejection. To address these issues, various antifouling strategies have been extensively studied, including polyethylene glycol-based polymer brushes. Conducting polymers-based biointerfaces have emerged as advanced surfaces for interfacing biological tissues and organs with electronics. Antifouling of such biointerfaces is a challenge. In this study, we fabricated electrospun fibre mats from sulphonated polystyrene-block-poly(ethylene-ran-butylene)-block-polystyrene (sSEBS), infused with conducting polymer poly(3,4-ethylenedioxythiophene) (PEDOT) (sSEBS-PEDOT), to produce a conductive (2.06 ± 0.1 S/cm), highly porous, fibre mat that can be used as a biointerface in bioelectronic applications. To afford antifouling, here the poly(oligo (ethylene glycol) methyl ether methacrylate) (POEGMA) brushes were grafted onto the sSEBS-PEDOT conducting fibre mats via surface-initiated atom transfer radical polymerization technique (SI-ATRP). For that, a copolymer of EDOT and an EDOT derivative with SI-ATRP initiating sites, 3,4-ethylenedioxythiophene) methyl 2-bromopropanoate (EDOTBr), was firstly electropolymerized on the sSEBS-PEDOT fibre mat to provide sSEBS-PEDOT/P(EDOT-co-EDOTBr). The POEGMA brushes were grafted from the sSEBS-PEDOT/P(EDOT-co-EDOTBr) and the polymerization kinetics confirmed the successful growth of the brushes. Fibre mats with 10-mers and 30-mers POEGMA brushes were studied for antifouling using a BCA protein assay. The mats with 30-mers grafted brushes exhibited excellent antifouling efficiency, ~82% of proteins repelled, compared to the pristine sSEBS-PEDOT fibre mat. The grafted fibre mats exhibited cell viability >80%, comparable to the standard cell culture plate controls. Such conducting, porous biointerfaces with POEGMA grafted brushes are suitable for applications in various biomedical devices, including biosensors, liquid biopsy, wound healing substrates and drug delivery systems.
Collapse
Affiliation(s)
- Jesna Ashraf
- Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand
- The MacDiarmid Institute of Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Sandy Lau
- Hub for Extracellular Vesicles Investigation (HEVI), Department of Obstetrics and Gynecology, The University of Auckland, Auckland 1010, New Zealand
| | - Alireza Akbarinejad
- Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand
- The MacDiarmid Institute of Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Clive W. Evans
- Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - David E. Williams
- Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand
- The MacDiarmid Institute of Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - David Barker
- Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand
- The MacDiarmid Institute of Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Jadranka Travas-Sejdic
- Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand
- The MacDiarmid Institute of Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| |
Collapse
|
20
|
Xie Y, Liang H, Jiang N, Liu D, Zhang N, Li Q, Zhang K, Sang X, Feng Y, Chen R, Zhang Y, Chen Q. Graphene quantum dots induce cascadic apoptosis via interaction with proteins associated with anti-oxidation after endocytosis by Trypanosoma brucei. Front Immunol 2022; 13:1022050. [PMID: 36561761 PMCID: PMC9763322 DOI: 10.3389/fimmu.2022.1022050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Trypanosoma brucei, the pathogen causing African sleeping sickness (trypanosomiasis) in humans, causes debilitating diseases in many regions of the world, but mainly in African countries with tropical and subtropical climates. Enormous efforts have been devoted to controlling trypanosomiasis, including expanding vector control programs, searching for novel anti-trypanosomial agents, and developing vaccines, but with limited success. In this study, we systematically investigated the effect of graphene quantum dots (GQDs) on trypanosomal parasites and their underlying mechanisms. Ultrasmall-sized GQDs can be efficiently endocytosed by T. brucei and with no toxicity to mammalian-derived cells, triggering a cascade of apoptotic reactions, including mitochondrial disorder, intracellular reactive oxygen species (ROS) elevation, Ca2+ accumulation, DNA fragmentation, adenosine triphosphate (ATP) synthesis impairment, and cell cycle arrest. All of these were caused by the direct interaction between GQDs and the proteins associated with cell apoptosis and anti-oxidation responses, such as trypanothione reductase (TryR), a key protein in anti-oxidation. GQDs specifically inhibited the enzymatic activity of TryR, leading to a reduction in the antioxidant capacity and, ultimately, parasite apoptotic death. These data, for the first time, provide a basis for the exploration of GQDs in the development of anti-trypanosomials.
Collapse
Affiliation(s)
- Yiwei Xie
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Hongrui Liang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Dingyuan Liu
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Naiwen Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Qilong Li
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Kai Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Yiwei Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China,*Correspondence: Qijun Chen,
| |
Collapse
|
21
|
Comprehensive Assessment of Antioxidant and Anti-Inflammatory Properties of Papaya Extracts. Foods 2022; 11:3211. [PMCID: PMC9601897 DOI: 10.3390/foods11203211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Antioxidant and anti-inflammatory properties of papaya (Carica papaya) fruits were evaluated to provide comprehensive information associated with the bioactive compounds. ‘Tainung No. 2’ papaya fruits, cultivated in a greenhouse, Korea, were harvested at unripe and ripe stages and then divided into seed and peel-pulp. Total phenolic and flavonoid contents were determined using spectrophotometry, and individual phenolic compounds were relatively quantified by HPLC-DAD and fifteen standards. Antioxidant activities were measured using four assays: DPPH (2,2-diphenyl-1-picrylhydrazyl) and ABTS (2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid)) scavenging activities, inhibition of lipid peroxidation, and FRAP (ferric reducing antioxidant power). Anti-inflammatory activities were measured by the regulation of NF-κB signaling pathways with the measurements of ROS and NO productions as the degree of oxidative stress. Total phenol contents increased in seed and peel–pulp extracts during ripening; flavonoid contents increased only in seed extracts. Total phenolic contents were associated with ABTS radical scavenging activity and FRAP. Of fifteen phenolic compounds, chlorogenic acid, cynarin, eupatorine, neochlorogenic acid, and vicenin II were identified among papaya extracts. ROS and NO productions were inhibited in papaya extracts. Especially, NO productions were inhibited higher in ripe seed extracts than in other extracts, which would be associated with the suppression of NF-κB activation and iNOS expression. These results suggest that papaya fruit extracts, including seeds, peels, and pulps, could be potential raw materials for functional foods.
Collapse
|
22
|
Cetin Aluc C, Gok B, Kecel-Gunduz S, Budama-Kilinc Y. Glycyrrhizic acid Poly(D,L-lactide-co-glycolide) nanoparticles: anti-aging cosmeceutical formulation for topical applications. PeerJ 2022. [DOI: 10.7717/peerj.14139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Glycyrrhizic acid (GA) is one of the components of licorice roots (Glycyrrhiza glabra L.). GA is a triterpenoid saponin can be used as a medicinal plant with its antiallergic, antiviral, anti-inflammatory, anti-ulcer, hepatoprotective, anticancer, anti-oxidation activities and several other therapeutic properties. The aim of this study is to develop an anti-aging formulation for topical application containing GA. In this context, GA-loaded Poly (D,L-lactide-co-glycolide) (PLGA) nanoparticles (NPs) were prepared using the double emulsion method, and were characterized by various spectroscopic methods. The efficacy of GA-PLGA NPs was evaluated with in vitro and in silico methods. The encapsulation efficiency and loading capacity were calculated. The in vitro release study was conducted, and the GA release profile was determined. The genotoxic activity of GA and GA-PLGA NPs was evaluated by the Ames test using TA98 and TA100 mutant strains of Salmonella typhimurium. The cytotoxic potential of GA-PLGA NPs was evaluated on the HaCaT cell line using the MTT assay. According to the genotoxicity and cytotoxicity results, it was found that the GA-PLGA NP formulation did not exhibit genotoxic and cytotoxic effects. Moreover, the efficacy of GA in preventing UVB-induced photo-aging in HaCaT cells and the clarification of the molecular mechanism of GA binding to MMPs were revealed by molecular docking analysis. In addition, through molecular dynamics (MD) analysis, the binding interaction of GA with MMPs in a dynamic system, and protein-ligand stability were predicted as a result of 50 ns MD simulation studies considering various analysis parameters. Finally, it was evaluated that GA-PLGA nanoformulation might be used as an alternative anti-aging skin care product candidate via topical application.
Collapse
Affiliation(s)
- Cigdem Cetin Aluc
- Graduate School of Natural and Applied Science, Yildiz Technical University, Istanbul, Türkiye
- Abdi Ibrahim Pharmaceuticals, Abdi Ibrahim Production Facilities, Istanbul, Türkiye
| | - Bahar Gok
- Graduate School of Natural and Applied Science, Yildiz Technical University, Istanbul, Türkiye
| | | | | |
Collapse
|
23
|
Manai F, Amadio M. Dimethyl Fumarate Triggers the Antioxidant Defense System in Human Retinal Endothelial Cells through Nrf2 Activation. Antioxidants (Basel) 2022; 11:antiox11101924. [PMID: 36290650 PMCID: PMC9598343 DOI: 10.3390/antiox11101924] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 12/06/2022] Open
Abstract
Dimethyl fumarate (DMF) is a well-known activator of Nrf2 (NF-E2-related factor 2), used in the treatment of psoriasis and multiple sclerosis. The mechanism of action consists in the modification of the cysteine residues on the Nrf2-inhibitor Keap1, thus leading to the dissociation of these two proteins and the consequent activation of Nrf2. Considering the paucity of evidence of DMF effects in the context of retinal endothelium, this in vitro study investigated the role of DMF in human retinal endothelial cells (HREC). Here, we show for the first time in HREC that DMF activates the Nrf2 pathway, thus leading to an increase in HO-1 protein levels and a decrease in intracellular ROS levels. Furthermore, this molecule also shows beneficial properties in a model of hyperglucose stress, exerting cytoprotective prosurvival effects. The overall collected results suggest that DMF-mediated activation of the Nrf2 pathway may also be a promising strategy in ocular diseases characterized by oxidative stress. This study opens a new perspective on DMF and suggests its potential repositioning in a broader therapeutical context.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Marialaura Amadio
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
- Correspondence: ; Tel.: +39-0382-987888
| |
Collapse
|
24
|
Cheng S, Wu D, Yuan L, Liu H, Ei-Seedi HR, Du M. Crassostrea gigas-Based Bioactive Peptide Protected Thrombin-Treated Endothelial Cells against Thrombosis and Cell Barrier Dysfunction. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:9664-9673. [PMID: 35900011 DOI: 10.1021/acs.jafc.2c02435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The activation of thrombin-treated endothelial cells resulted in disruption of the vascular tissues. A novel oyster-derived bioactive dodecapeptide (IEELEELEAER, P-2-CG) was reported to protect the human umbilical vein endothelial cells and their barrier function via the decrease of VE-cadherin disruption and the restoration of the F-actin arrangement. The promotion of the extrinsic pathway in this case triggers the release of tissue factors that occurs on the surface of the endothelial cells, thus changing the antithrombotic to prothrombotic. P-2-CG induced accordingly a prolongation of plasma clotting time and thrombin generation time, following the alteration of the antithrombotic phenotype. Furthermore, the antithrombotic activity of P-2-CG was also supported by the reduction of FXa and the inhibition of other factors release, for instance, inflammation factors, ROS, etc. In addition to its antithrombogenic role, P-2-CG displayed anti-inflammatory and antioxidant properties via the mitogen-activated protein kinase cascades and central signaling pathways as shown in an in vitro model of endothelial dysfunction.
Collapse
Affiliation(s)
- Shuzhen Cheng
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Di Wu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Lushun Yuan
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Nephrology, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Hanxiong Liu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Hesham R Ei-Seedi
- Pharmacognosy Group, Department of Medicinal Chemistry, Uppsala University, Biomedical Centre, Uppsala 75123, Sweden
| | - Ming Du
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
25
|
Alhussan A, Palmerley N, Smazynski J, Karasinska J, Renouf DJ, Schaeffer DF, Beckham W, Alexander AS, Chithrani DB. Potential of Gold Nanoparticle in Current Radiotherapy Using a Co-Culture Model of Cancer Cells and Cancer Associated Fibroblast Cells. Cancers (Basel) 2022; 14:cancers14153586. [PMID: 35892845 PMCID: PMC9332249 DOI: 10.3390/cancers14153586] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Many cancer therapeutics do not account for the complexity of the tumor microenvironment (TME), which may result in failure when applied clinically. In this paper we utilized a simple tumor model made of two types of pancreatic cancer cells that contribute to the tumor environment, i.e., cancer cells and cancer associated fibroblasts. Herein, radiotherapy along with radiosensitizing gold nanoparticles were used to test the efficacy of a co-culture vs. monoculture model. The results show that the co-culture model exhibited heightened resistance to radiation. Furthermore, we found that the combination of gold radiosensitizers with radiotherapy reduced the radioresistance of the co-culture model compared to radiotherapy alone. This study demonstrates the potential of using nanotherapeutics in targeting the complex tumor microenvironment. Abstract Many cancer therapeutics are tested in vitro using only tumour cells. However, the tumour promoting effect of cancer associated fibroblasts (CAFs) within the tumour microenvironment (TME) is thought to reduce cancer therapeutics’ efficacy. We have chosen pancreatic ductal adenocarcinoma (PDAC) as our tumor model. Our goal is to create a co-culture of CAFs and tumour cells to model the interaction between cancer and stromal cells in the TME and allow for better testing of therapeutic combinations. To test the proposed co-culture model, a gold nanoparticle (GNP) mediated-radiation response was used. Cells were grown in co-culture with different ratios of CAFs to cancer cells. MIA PaCa-2 was used as our PDAC cancer cell line. Co-cultured cells were treated with 2 Gy of radiation following GNP incubation. DNA damage and cell proliferation were examined to assess the combined effect of radiation and GNPs. Cancer cells in co-culture exhibited up to a 23% decrease in DNA double strand breaks (DSB) and up to a 35% increase in proliferation compared to monocultures. GNP/Radiotherapy (RT) induced up to a 25% increase in DNA DSBs and up to a 15% decrease in proliferation compared to RT alone in both monocultured and co-cultured cells. The observed resistance in the co-culture system may be attributed to the role of CAFs in supporting cancer cells. Moreover, we were able to reduce the activity of CAFs using GNPs during radiation treatment. Indeed, CAFs internalize a significantly higher number of GNPs, which may have led to the reduction in their activity. One reason experimental therapeutics fail in clinical trials relates to limitations in the pre-clinical models that lack a true representation of the TME. We have demonstrated a co-culture platform to test GNP/RT in a clinically relevant environment.
Collapse
Affiliation(s)
- Abdulaziz Alhussan
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.A.); (N.P.); (W.B.)
| | - Nicholas Palmerley
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.A.); (N.P.); (W.B.)
| | - Julian Smazynski
- Deeley Research Centre, British Columbia Cancer—Victoria, Victoria, BC V8R 6V5, Canada;
| | - Joanna Karasinska
- Pancreas Centre BC, Vancouver, BC V5Z 1G1, Canada; (J.K.); (D.J.R.); (D.F.S.)
| | - Daniel J. Renouf
- Pancreas Centre BC, Vancouver, BC V5Z 1G1, Canada; (J.K.); (D.J.R.); (D.F.S.)
| | - David F. Schaeffer
- Pancreas Centre BC, Vancouver, BC V5Z 1G1, Canada; (J.K.); (D.J.R.); (D.F.S.)
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - Wayne Beckham
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.A.); (N.P.); (W.B.)
- Radiation Oncology, British Columbia Cancer—Victoria, Victoria, BC V8R 6V5, Canada;
| | - Abraham S. Alexander
- Radiation Oncology, British Columbia Cancer—Victoria, Victoria, BC V8R 6V5, Canada;
| | - Devika B. Chithrani
- Department of Physics and Astronomy, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.A.); (N.P.); (W.B.)
- Radiation Oncology, British Columbia Cancer—Victoria, Victoria, BC V8R 6V5, Canada;
- Centre for Advanced Materials and Related Technologies, Department of Chemistry, University of Victoria, Victoria, BC V8P 5C2, Canada
- Centre for Biomedical Research, Department of Biology, University of Victoria, Victoria, BC V8P 5C2, Canada
- Department of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
- Department of Computer Science, Mathematics, Physics and Statistics, Okanagan Campus, University of British Columbia, Kelowna, BC V1V 1V7, Canada
- Correspondence:
| |
Collapse
|
26
|
Moesin Serves as Scaffold Protein for PD-L1 in Human Uterine Cervical Squamous Carcinoma Cells. J Clin Med 2022; 11:jcm11133830. [PMID: 35807113 PMCID: PMC9267616 DOI: 10.3390/jcm11133830] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/17/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapy targeting the programmed death ligand-1 (PD-L1)/PD-1 axis has emerged as a promising treatment for uterine cervical cancer; however, only a small subset of patients with uterine cervical squamous cell carcinoma (SCC) derives clinical benefit from ICB therapies. Thus, there is an urgent unmet medical need for novel therapeutic strategies to block the PD-L1/PD-1 axis in patients with uterine cervical SCC. Here, we investigated the involvement of ezrin/radixin/moesin (ERM) family scaffold proteins, which crosslink several plasma membrane proteins with the actin cytoskeleton, on the plasma membrane localization of PD-L1 in BOKU and HCS-2 cells derived from human uterine cervical SCC. Immunofluorescence analysis showed that PD-L1 colocalized with all three ERM proteins in the plasma membrane. Gene knockdown of moesin, but not ezrin and radixin, substantially reduced the plasma membrane expression of PD-L1, with limited effect on mRNA expression. An immunoprecipitation assay demonstrated the molecular interaction between PD-L1 and moesin. Moreover, phosphorylated, i.e., activated, moesin was highly colocalized with PD-L1 in the plasma membrane. In conclusion, moesin may be a scaffold protein responsible for the plasma membrane expression of PD-L1 in human uterine cervical SCC.
Collapse
|
27
|
Olivares-Galván S, Marina M, García M. Extraction of valuable compounds from brewing residues: Malt rootlets, spent hops, and spent yeast. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
28
|
Luzak B, Siarkiewicz P, Boncler M. An evaluation of a new high-sensitivity PrestoBlue assay for measuring cell viability and drug cytotoxicity using EA.hy926 endothelial cells. Toxicol In Vitro 2022; 83:105407. [PMID: 35659575 DOI: 10.1016/j.tiv.2022.105407] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/29/2022] [Accepted: 05/29/2022] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Commercially-available resazurin-based reagents used for cell viability assessment contain varying amounts of resorufin; these may contribute to differences in autofluorescence, signal-to-background (S/B) ratio and the dynamic range of the assay. OBJECTIVES This in vitro study compares the sensitivity of a new, high-sensitivity PrestoBlue (hs-PB) assay with standard PrestoBlue (PB) in assessing the efficacy of valinomycin and antimycin A in human vascular endothelial EA.hy926 cells, as well as cell viability. METHODS The metabolic activity of EA.hy926 was evaluated based on resorufin fluorescence or formazan absorbance. RESULTS The hs-PB assay demonstrated lower resorufin autofluorescence than the PB, resulting in a ≥ 1.4-fold increase in S/B ratio in hs-PB compared to PB. Valinomycin was more potent cytotoxic agent than antimycin A. The hs-PB, PB and MTT produced similar IC50 values for valinomycin. Antimycin A demonstrated significantly higher potency in the MTT than in the resazurin-based assays. The EA.hy926 cells demonstrated higher metabolic activity in the presence of the antimycin A solvent - DMSO. CONCLUSION All the examined methods may be used interchangeably to analyze drug cytotoxicity. Any differences in drug cytotoxicity observed between the assays may be due to relatively low drug potency and/or the influence of solvent on metabolism of assay reagent. The hs-PB assay appears to more effectively detect cell viability and produce a stronger signal than its conventional counterpart.
Collapse
Affiliation(s)
- Boguslawa Luzak
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland
| | - Przemysław Siarkiewicz
- Institute of Polymer and Dye Technology, Faculty of Chemistry, Lodz University of Technology, Lodz, Poland
| | - Magdalena Boncler
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
29
|
Chen J, Li J, Fan T, Zhong S, Qin X, Li R, Gao J, Liang Y. Protective effects of curcumin/cyclodextrin polymer inclusion complex against hydrogen peroxide-induced LO2 cells damage. Food Sci Nutr 2022; 10:1649-1656. [PMID: 35592280 PMCID: PMC9094476 DOI: 10.1002/fsn3.2787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/11/2022] [Accepted: 01/29/2022] [Indexed: 11/12/2022] Open
Abstract
The objective of the present study was to explore the protective effects of the curcumin/cyclodextrin polymer (CUR/CDP) inclusion complex on hydrogen peroxide (H2O2)-induced LO2 cells damage. In this study, a H2O2-induced cells oxidative injury model was established to test the protective effects of the CUR/CDP inclusion complex. The cell viability of cells was detected by the thiazolyl blue tetrazolium bromide (MTT) assay. The extracellular lactate dehydrogenase (LDH) activity, catalase (CAT) activity, and malondialdehyde (MDA) level were detected by assay kits. The cellular reactive oxygen species (ROS) level was detected using the dichlorodihydrofluorescein (DCF) fluorescence assay. Western blotting analysis was conducted to assess the changes of phosphorylated-p53 and caspase-3. The results showed that 700 μM H2O2-treated LO2 cells for 3 h resulted in a significant decrease of cell viability to 53.00 ± 1.68%, which established the cell oxidative injury model. Cells treated with H2O2 led to a significant increase of extracellular LDH activity, MDA content, and ROS level, and decreased CAT activity. Treatment with CUR/CDP significantly reversed the changes of the above indicators. Moreover, CUR/CDP treatment at 20 and 40 μg/ml inhibited H2O2-induced increase in phosphorylated-p53 and caspase-3 expression, indicating that CUR/CDP suppressed cell apoptosis to alleviate liver injury. The results of those studies demonstrated that CUR/CDP had a protective effect on the oxidative damage of LO2 cells, and it could be developed as a new type of natural liver protection product to apply in the prevention of liver injury.
Collapse
Affiliation(s)
- Jianping Chen
- College of Food Science and TechnologyGuangdong Provincial Key Laboratory of Aquatic Product Processing and SafetyGuangdong Provincial Engineering Technology Research Center of SeafoodGuangdong Province Engineering Laboratory for Marine Biological ProductsKey Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education InstitutionGuangdong Ocean UniversityZhanjiangChina
- Collaborative Innovation Center of Seafood Deep ProcessingDalian Polytechnic UniversityDalianChina
| | - Jiarui Li
- College of Food Science and TechnologyGuangdong Provincial Key Laboratory of Aquatic Product Processing and SafetyGuangdong Provincial Engineering Technology Research Center of SeafoodGuangdong Province Engineering Laboratory for Marine Biological ProductsKey Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education InstitutionGuangdong Ocean UniversityZhanjiangChina
- Collaborative Innovation Center of Seafood Deep ProcessingDalian Polytechnic UniversityDalianChina
| | - Tugui Fan
- College of Food Science and TechnologyGuangdong Provincial Key Laboratory of Aquatic Product Processing and SafetyGuangdong Provincial Engineering Technology Research Center of SeafoodGuangdong Province Engineering Laboratory for Marine Biological ProductsKey Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education InstitutionGuangdong Ocean UniversityZhanjiangChina
- Collaborative Innovation Center of Seafood Deep ProcessingDalian Polytechnic UniversityDalianChina
| | - Saiyi Zhong
- College of Food Science and TechnologyGuangdong Provincial Key Laboratory of Aquatic Product Processing and SafetyGuangdong Provincial Engineering Technology Research Center of SeafoodGuangdong Province Engineering Laboratory for Marine Biological ProductsKey Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education InstitutionGuangdong Ocean UniversityZhanjiangChina
- Collaborative Innovation Center of Seafood Deep ProcessingDalian Polytechnic UniversityDalianChina
| | - Xiaoming Qin
- College of Food Science and TechnologyGuangdong Provincial Key Laboratory of Aquatic Product Processing and SafetyGuangdong Provincial Engineering Technology Research Center of SeafoodGuangdong Province Engineering Laboratory for Marine Biological ProductsKey Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education InstitutionGuangdong Ocean UniversityZhanjiangChina
- Collaborative Innovation Center of Seafood Deep ProcessingDalian Polytechnic UniversityDalianChina
| | - Rui Li
- College of Food Science and TechnologyGuangdong Provincial Key Laboratory of Aquatic Product Processing and SafetyGuangdong Provincial Engineering Technology Research Center of SeafoodGuangdong Province Engineering Laboratory for Marine Biological ProductsKey Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education InstitutionGuangdong Ocean UniversityZhanjiangChina
- Collaborative Innovation Center of Seafood Deep ProcessingDalian Polytechnic UniversityDalianChina
| | - Jialong Gao
- College of Food Science and TechnologyGuangdong Provincial Key Laboratory of Aquatic Product Processing and SafetyGuangdong Provincial Engineering Technology Research Center of SeafoodGuangdong Province Engineering Laboratory for Marine Biological ProductsKey Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education InstitutionGuangdong Ocean UniversityZhanjiangChina
- Collaborative Innovation Center of Seafood Deep ProcessingDalian Polytechnic UniversityDalianChina
| | - Yuanwei Liang
- College of Chemistry and EnvironmentGuangdong Ocean UniversityZhanjiangChina
| |
Collapse
|
30
|
Vieira AM, Silvestre OF, Silva BF, Ferreira CJ, Lopes I, Gomes AC, Espiña B, Sárria MP. pH-sensitive nanoliposomes for passive and CXCR-4-mediated marine yessotoxin delivery for cancer therapy. Nanomedicine (Lond) 2022; 17:717-739. [PMID: 35481356 DOI: 10.2217/nnm-2022-0010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Yessotoxin (YTX), a marine-derived drug, was encapsulated in PEGylated pH-sensitive nanoliposomes, covalently functionalized (strategy I) with SDF-1α and by nonspecific adsorption (strategy II), to actively target chemokine receptor CXCR-4. Methods: Cytotoxicity to normal human epithelial cells (HK-2) and prostate (PC-3) and breast (MCF-7) adenocarcinoma models, with different expression levels of CXCR-4, were tested. Results: Strategy II exerted the highest cytotoxicity toward cancer cells while protecting normal epithelia. Acid pH-induced fusion of nanoliposomes seemed to serve as a primary route of entry into MCF-7 cells but PC-3 data support an endocytic pathway for their internalization. Conclusion: This work describes an innovative hallmark in the current marine drug clinical pipeline, as the developed nanoliposomes are promising candidates in the design of groundbreaking marine flora-derived anticancer nanoagents.
Collapse
Affiliation(s)
- Ana Mg Vieira
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal.,Centre of Molecular & Environmental Biology (CBMA), University of Minho, Braga, 4710-057, Portugal
| | - Oscar F Silvestre
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal
| | - Bruno Fb Silva
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal
| | - Celso Jo Ferreira
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal.,Centro de Física das Universidades do Minho e do Porto (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
| | - Ivo Lopes
- Centre of Molecular & Environmental Biology (CBMA), University of Minho, Braga, 4710-057, Portugal
| | - Andreia C Gomes
- Centre of Molecular & Environmental Biology (CBMA), University of Minho, Braga, 4710-057, Portugal.,Institute of Science & Innovation for Biosustainability (IB-S), University of Minho, Braga, 4710-057, Portugal
| | - Begoña Espiña
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal
| | - Marisa P Sárria
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga, 4715-330, Portugal
| |
Collapse
|
31
|
Tameishi M, Ishikawa H, Tanaka C, Kobori T, Urashima Y, Ito T, Obata T. Ezrin Contributes to the Plasma Membrane Expression of PD-L1 in A2780 Cells. J Clin Med 2022; 11:jcm11092457. [PMID: 35566582 PMCID: PMC9100183 DOI: 10.3390/jcm11092457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 01/30/2023] Open
Abstract
Programmed death ligand–1 (PD–L1) is one of the immune checkpoint molecule localized on the plasma membrane of numerous cancer cells that negatively regulates T-cell-mediated immunosurveillance. Despite the remarkable efficacy and safety profile of immune checkpoint inhibitors (ICIs), such as anti-PD–L1 antibodies, restricted poor therapeutic responses to ICIs are often observed in patients with ovarian cancer. Because higher expression of PD–L1 in advanced ovarian cancer is associated with a decreased survival rate, identifying the potential molecules to regulate the plasma membrane expression of PD–L1 may provide a novel therapeutic strategy to improve the efficacy of ICIs against ovarian cancers. Here, we reveal the involvement of the ezrin/radixin/moesin (ERM) family, which crosslinks transmembrane proteins with the actin cytoskeleton by serving as a scaffold protein, in the plasma membrane expression of PD–L1 in the human epithelial ovarian cancer cell line A2780. Our results demonstrate that PD–L1 and all three ERMs were expressed at the mRNA and protein levels in A2780 cells, and that PD–L1 was highly colocalized with ezrin and moesin, but moderately with radixin, in the plasma membrane. Interestingly, RNA interference-mediated gene silencing of ezrin, but not of radixin or moesin, substantially reduced the plasma membrane expression of PD–L1 without altering its mRNA expression. In conclusion, our results indicate that ezrin may be responsible for the plasma membrane expression of PD–L1, possibly by serving as a scaffold protein in A2780 cells. Ezrin is a potential therapeutic target for improving the efficacy of ICIs against ovarian cancers.
Collapse
Affiliation(s)
- Mayuka Tameishi
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
| | - Honami Ishikawa
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
| | - Chihiro Tanaka
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
| | - Takuro Kobori
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
| | - Yoko Urashima
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
| | - Takuya Ito
- Laboratory of Natural Medicines, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan;
| | - Tokio Obata
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
- Correspondence: ; Tel.: +81-721-24-9371
| |
Collapse
|
32
|
Yazdanpanah Z, Johnston JD, Cooper DML, Chen X. 3D Bioprinted Scaffolds for Bone Tissue Engineering: State-Of-The-Art and Emerging Technologies. Front Bioeng Biotechnol 2022; 10:824156. [PMID: 35480972 PMCID: PMC9035802 DOI: 10.3389/fbioe.2022.824156] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Treating large bone defects, known as critical-sized defects (CSDs), is challenging because they are not spontaneously healed by the patient’s body. Due to the limitations associated with conventional bone grafts, bone tissue engineering (BTE), based on three-dimensional (3D) bioprinted scaffolds, has emerged as a promising approach for bone reconstitution and treatment. Bioprinting technology allows for incorporation of living cells and/or growth factors into scaffolds aiming to mimic the structure and properties of the native bone. To date, a wide range of biomaterials (either natural or synthetic polymers), as well as various cells and growth factors, have been explored for use in scaffold bioprinting. However, a key challenge that remains is the fabrication of scaffolds that meet structure, mechanical, and osteoconductive requirements of native bone and support vascularization. In this review, we briefly present the latest developments and discoveries of CSD treatment by means of bioprinted scaffolds, with a focus on the biomaterials, cells, and growth factors for formulating bioinks and their bioprinting techniques. Promising state-of-the-art pathways or strategies recently developed for bioprinting bone scaffolds are highlighted, including the incorporation of bioactive ceramics to create composite scaffolds, the use of advanced bioprinting technologies (e.g., core/shell bioprinting) to form hybrid scaffolds or systems, as well as the rigorous design of scaffolds by taking into account of the influence of such parameters as scaffold pore geometry and porosity. We also review in-vitro assays and in-vivo models to track bone regeneration, followed by a discussion of current limitations associated with 3D bioprinting technologies for BTE. We conclude this review with emerging approaches in this field, including the development of gradient scaffolds, four-dimensional (4D) printing technology via smart materials, organoids, and cell aggregates/spheroids along with future avenues for related BTE.
Collapse
Affiliation(s)
- Zahra Yazdanpanah
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- *Correspondence: Zahra Yazdanpanah,
| | - James D. Johnston
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - David M. L. Cooper
- Department of Anatomy Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
33
|
Dong P, Zhan Y, Jusuf S, Hui J, Dagher Z, Mansour MK, Cheng J. Photoinactivation of Catalase Sensitizes Candida albicans and Candida auris to ROS-Producing Agents and Immune Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104384. [PMID: 35119220 PMCID: PMC8981478 DOI: 10.1002/advs.202104384] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/07/2022] [Indexed: 06/14/2023]
Abstract
Microbes have developed their own specific strategies to cope with reactive oxygen species (ROS). Catalase, a heme-containing tetramer expressed in a broad range of aerobic fungi, shows remarkable efficiency in degrading hydrogen peroxide (H2 O2 ) for fungal survival and host invasion. Here, it is demonstrated that catalase inactivation by blue light renders fungal cells highly susceptible to ROS attack. To confirm catalase as a major molecular target of blue light, wild type Candida albicans are systematically compared with a catalase-deficient mutant strain regarding their susceptibility to ROS through 410 nm treatment. Upon testing a wide range of fungal species, it is found that intracellular catalase can be effectively and universally inactivated by 410 nm blue light. It is also found that photoinactivation of catalase in combination with ROS-generating agents is highly effective in total eradication of various fungal species, including multiple Candida auris strains, the causative agent of the global fungal epidemic. In addition, photoinactivation of catalase is shown to facilitate macrophage killing of intracellular Candida albicans. The antifungal efficacy of catalase photoinactivation is further validated using a C. albicans-induced mouse model of skin abrasion. Taken together, the findings offer a novel catalase-photoinactivation approach to address multidrug-resistant Candida infections.
Collapse
Affiliation(s)
- Pu‐Ting Dong
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Photonics CenterBoston UniversityBostonMA02215USA
| | - Yuewei Zhan
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Photonics CenterBoston UniversityBostonMA02215USA
| | - Sebastian Jusuf
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Photonics CenterBoston UniversityBostonMA02215USA
| | - Jie Hui
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Photonics CenterBoston UniversityBostonMA02215USA
| | - Zeina Dagher
- Division of Infectious DiseasesMassachusetts General HospitalBostonMA02114USA
- Harvard Medical SchoolBostonMA02115USA
| | - Michael K. Mansour
- Division of Infectious DiseasesMassachusetts General HospitalBostonMA02114USA
- Harvard Medical SchoolBostonMA02115USA
| | - Ji‐Xin Cheng
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Photonics CenterBoston UniversityBostonMA02215USA
| |
Collapse
|
34
|
Abhijit Saha, Sarker K, Ghosh A, Mishra S, Sen S. Analogue Based Design, Synthesis, Biological Evaluation, and Molecular Docking of Some Thalidomide Metabolites as Selective Cytotoxic and Antiangiogenic Agents against Multiple Myeloma. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
35
|
|
36
|
Priya VT, N B, V S, P S, N D K, C K, Alfarhan A, Antonisamy P. Partially purified lead molecules from Dodonaea viscosa and their antimicrobial efficacy against infectious human pathogens. J Infect Public Health 2021; 14:1822-1830. [PMID: 34836798 DOI: 10.1016/j.jiph.2021.11.007] [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: 08/24/2021] [Revised: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND The utilization of medicinal plants and their drugs have the advantage of reducing side effects compared with manufactured antimicrobials. Artificial drugs have unpleasant side effects, further, the number of drug resistant pathogens is increasing, thus huge challenge for control of resistant pathogens. Therefore, the current research explores the potential of partially purified bioactive compounds from Dodonaea viscosa against certain human pathogens. METHODS Healthy leaves of D. viscosa (L.) were collected, extracted and optimized with different solvents. Preliminary phytochemical screening of the extracts was done and antibacterial activities were tested against human pathogens. The active crude extract was further purified by column chromatography and the homogeneity was confirmed by thin layer chromatography (TLC). The partially purified compounds were screened further for antibacterial, antibiofilm and anticancer activities. RESULTS The crude ethanol extract of D. viscosa leaves showed the presence of phytochemical like tannins, alkaloids, flavanoids, terpenoids, glycosides, steroids and phenols. Ethanol extract exhibited the maximum zone of inhibition (11 mm) against S. agalactiae, B. cereus, S. typhi and E. coli at 15 mg when compared with other bacteria. Column chromatography fractions Dv12 and Dv20 exhibited the maximum zone of inhibition against B. cereus. 1000 μg of Dv12 partially purified compound against streptococcus isolates in glass test tube showed biofilm inhibition range of 34.4%-63.1%. Whereas B. cereus, S. aureus, S. typhi, and K. pneumoniae showed 31.1%-53.6% biofilm inhibition compared to curcumin control. Active fractions of Dv12 and Dv20 increased concentration confirmed that the gradual decrease in cell density and possesses growth inhibition towards A 549 human lung adenocarcinoma cells. CONCLUSION We have extracted the bioactive compounds from D. viscosa (L.) leaves and tested the activity of a partially purified compound against human pathogenic bacteria, biofilm formation and cytotoxicity against A 549 human lung adenocarcinoma cells. The purified bioactive compounds might be used as therapeutic agents against different microbial infections such as skin infection, throat infection and other infectious diseases.
Collapse
Affiliation(s)
- V Thamil Priya
- Department of Chemistry, VHNSN College, Virudhunagar, 626 001, Tamil Nadu, India
| | - Balasubramanian N
- Department of Immunology, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625 021, Tamil Nadu, India
| | - Shanmugaiah V
- Department of Microbial Technology, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625 021, Tamil Nadu, India.
| | - Sathishkumar P
- Department of Plant Biotechnology, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, Tamil Nadu, India
| | - Kannan N D
- Department of Plant Biotechnology, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, Tamil Nadu, India
| | - Karunakaran C
- Department of Chemistry, VHNSN College, Virudhunagar, 626 001, Tamil Nadu, India.
| | - Ahmed Alfarhan
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box-2455, Riyadh 11451, Saudi Arabia
| | - Paulrayer Antonisamy
- Ilwonbio Co., Ltd., Department of Physiology, Wonkwang University School of Korean Medicine, Iksan, Jeonbuk, South Korea
| |
Collapse
|
37
|
Zehra S, Cirilli I, Silvestri S, Gómez-Ruiz S, Tabassum S, Arjmand F. Structure elucidation, in vitro binding studies and ROS-dependent anti-cancer activity of Cu(II) and Zn(II) phthaloylglycinate(phen) complexes against MDA-MB-231 cells. Metallomics 2021; 13:6415206. [PMID: 34724067 DOI: 10.1093/mtomcs/mfab064] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022]
Abstract
New mononuclear Cu(II) and Zn(II)-based complexes 1 [Cu(L)2(diimine)HOCH3] and 2 [Zn(L)2(diimine)] have been synthesized as anti-cancer chemotherapeutics targeted to tRNA. The structure elucidation of complexes 1 and 2 was carried out by spectroscopic and single X-ray diffraction studies. In vitro interaction studies of complexes 1 and 2 with ct-DNA/tRNA were performed by employing various biophysical techniques to evaluate and predict their interaction behavior and preferential selectivity at biomolecular therapeutic targets. The corroborative results of the interaction studies demonstrated that complexes 1 and 2 exhibited avid binding propensity via intercalative mode of binding toward ct-DNA/tRNA. Electrophoretic assay revealed that the complexes 1 and 2 were able to promote single- and double-strand cleavage of the plasmid DNA at low micromolar concentrations under physiological conditions in the absence of an additional oxidizing or reducing agent. RNA hydrolysis studies revealed that the complexes 1 and 2 could promote tRNA cleavage in a concentration and time-dependent manner. The cytotoxic potential of complexes 1 and 2 was evaluated against the MDA-MB-231 cell line, which showed that the complexes were able to inhibit the cell growth in a dose-dependent manner. The intracellular ROS production and mitochondrial superoxide anion assay revealed that the complexes 1 and 2 induce a dose-dependent activity, suggesting the involvement of ROS-mediated mitochondrial apoptotic pathway leading to cell death.
Collapse
Affiliation(s)
- Siffeen Zehra
- Department of Chemistry, Aligarh Muslim University, Aligarh, UP 202002, India
| | - Ilenia Cirilli
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy.,School of Pharmacy, University of Camerino, Camerino (MC) 62032, Italy
| | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Santiago Gómez-Ruiz
- COMET-NANO Group, Departamento de Biología y Geología, Física y Química Inorgánica,, E.S.C.E.T., Universidad Rey Juan Carlos, 28933 Móstoles, Madrid, Spain
| | - Sartaj Tabassum
- Department of Chemistry, Aligarh Muslim University, Aligarh, UP 202002, India
| | - Farukh Arjmand
- Department of Chemistry, Aligarh Muslim University, Aligarh, UP 202002, India
| |
Collapse
|
38
|
Costa MR, Gošar D, Pinti M, Ferreira A, Bergant Marušič M. In vitro toxicity of arsenic rich waters from an abandoned gold mine in northeast Portugal. ENVIRONMENTAL RESEARCH 2021; 202:111683. [PMID: 34270993 DOI: 10.1016/j.envres.2021.111683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/15/2021] [Accepted: 06/27/2021] [Indexed: 06/13/2023]
Abstract
This is a follow-up study of physicochemical water monitoring data from the abandoned Freixeda gold mine in Portugal, where arsenic (As) has remained above drinking water and irrigation limits over the years. The main objective of the current work was to investigate the toxicological potential of As-containing water on human cell line as an indicator of a potential health risk to humans. Six water samples collected in February 2018 were analysed for arsenic, major anions, cations and trace elements. Toxicity experiments were carried out on the human gastrointestinal cell line Caco-2 with five water samples containing As above 10 μg L-1. The results show that groundwater contains higher amounts of dissolved minerals than surface water, particularly with higher concentrations of SO42-, Fe and HCO3- and also higher As(III), reaching 336 μg L-1 (As(T) = 607 μg L-1). In surface waters As concentration decreased and reached 150 μg L-1, mainly as As(V). Metabolic activity was generally lower in Caco-2 cells exposed to As-containing water samples compared to pure As(III) solution, adapted to As concentrations, while production of reactive oxygen species (ROS) was higher. Short-term exposure to As-contaminated water samples also resulted in increased genotoxicity. This study suggests that mixture of As with various chemical elements in water may have a synergistic effect in promoting cytotoxicity. It is likely that prolonged exposure, as is common in areas with contaminated water, would have even more harmful effects.
Collapse
Affiliation(s)
- Maria R Costa
- Geology Department of UTAD, 5000-801, Vila Real, Portugal; GeoBioTec, Geosciences Department, University of Aveiro, Aveiro Santiago Campus, Portugal.
| | - Doroteja Gošar
- School of Environmental Sciences, University of Nova Gorica, Glavni trg 8, SI-5271, Vipava, Slovenia
| | - Marika Pinti
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Vipavska cesta 13, SI-5000, Nova Gorica, Slovenia
| | - Adelaide Ferreira
- LNEG - Laboratório Nacional de Energia e Geologia I.P., Rua da Amieira, Ap. 1089, 4466-901, S. Mamede de Infesta, Portugal.
| | - Martina Bergant Marušič
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Vipavska cesta 13, SI-5000, Nova Gorica, Slovenia.
| |
Collapse
|
39
|
Contribution of Ezrin on the Cell Surface Plasma Membrane Localization of Programmed Cell Death Ligand-1 in Human Choriocarcinoma JEG-3 Cells. Pharmaceuticals (Basel) 2021; 14:ph14100963. [PMID: 34681187 PMCID: PMC8540387 DOI: 10.3390/ph14100963] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/01/2021] [Accepted: 09/22/2021] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint blockade (ICB) antibodies targeting programmed cell death ligand-1 (PD-L1) and programmed cell death-1 (PD-1) have improved survival in patients with conventional single agent chemotherapy-resistant gestational trophoblastic neoplasia (GTN). However, many patients are resistant to ICB therapy, the mechanisms of which are poorly understood. Unraveling the regulatory mechanism for PD-L1 expression may provide a new strategy to improve ICB therapy in patients with GTN. Here, we investigated whether the ezrin/radixin/moesin (ERM) family, i.e., a group of scaffold proteins that crosslink actin cytoskeletons with several plasma membrane proteins, plays a role in the regulation of PD-L1 expression using JEG-3 cells, a representative human choriocarcinoma cell line. Our results demonstrate mRNA and protein expressions of ezrin, radixin, and PD-L1, as well as their colocalization in the plasma membrane. Intriguingly, immunoprecipitation experiments revealed that PD-L1 interacted with both ezrin and radixin and the actin cytoskeleton. Moreover, gene silencing of ezrin but not radixin strongly diminished the cell surface expression of PD-L1 without altering the mRNA level. These results indicate that ezrin may contribute to the cell surface localization of PD-L1 as a scaffold protein in JEG-3 cells, highlighting a potential therapeutic target to improve the current ICB therapy in GTN.
Collapse
|
40
|
Tanaka C, Kobori T, Tameishi M, Urashima Y, Ito T, Obata T. Ezrin Modulates the Cell Surface Expression of Programmed Cell Death Ligand-1 in Human Cervical Adenocarcinoma Cells. Molecules 2021; 26:5648. [PMID: 34577118 PMCID: PMC8469114 DOI: 10.3390/molecules26185648] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 01/01/2023] Open
Abstract
Cancer cells employ programmed cell death ligand-1 (PD-L1), an immune checkpoint protein that binds to programmed cell death-1 (PD-1) and is highly expressed in various cancers, including cervical carcinoma, to abolish T-cell-mediated immunosurveillance. Despite a key role of PD-L1 in various cancer cell types, the regulatory mechanism for PD-L1 expression is largely unknown. Understanding this mechanism could provide a novel strategy for cervical cancer therapy. Here, we investigated the influence of ezrin/radixin/moesin (ERM) family scaffold proteins, crosslinking the actin cytoskeleton and certain plasma membrane proteins, on the expression of PD-L1 in HeLa cells. Our results showed that all proteins were expressed at mRNA and protein levels and that all ERM proteins were highly colocalized with PD-L1 in the plasma membrane. Interestingly, immunoprecipitation assay results demonstrated that PD-L1 interacted with ERM as well as actin cytoskeleton proteins. Furthermore, gene silencing of ezrin, but not radixin and moesin, remarkably decreased the protein expression of PD-L1 without affecting its mRNA expression. In conclusion, ezrin may function as a scaffold protein for PD-L1; regulate PD-L1 protein expression, possibly via post-translational modification in HeLa cells; and serve as a potential therapeutic target for cervical cancer, improving the current immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Chihiro Tanaka
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Osaka, Japan; (C.T.); (M.T.); (Y.U.)
| | - Takuro Kobori
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Osaka, Japan; (C.T.); (M.T.); (Y.U.)
| | - Mayuka Tameishi
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Osaka, Japan; (C.T.); (M.T.); (Y.U.)
| | - Yoko Urashima
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Osaka, Japan; (C.T.); (M.T.); (Y.U.)
| | - Takuya Ito
- Laboratory of Natural Medicines, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Osaka, Japan;
| | - Tokio Obata
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Osaka, Japan; (C.T.); (M.T.); (Y.U.)
| |
Collapse
|
41
|
Muniyandi P, Palaninathan V, Mizuki T, Mohamed MS, Hanajiri T, Maekawa T. Scaffold mediated delivery of dual miRNAs to transdifferentiate cardiac fibroblasts. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112323. [PMID: 34474874 DOI: 10.1016/j.msec.2021.112323] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/30/2021] [Accepted: 07/12/2021] [Indexed: 01/14/2023]
Abstract
The standard scaffold-mediated delivery of drugs/biomolecules has been successful due to the unique attributes of scaffolds, specifically the electrospun polymeric scaffolds that mimic ECM are well suited for advanced regenerative applications. Cardiac tissue engineering includes the interpretation of cellular and molecular mechanisms concerning heart regeneration and identifying an efficient reprogramming strategy to overcome the limitation of delivery systems and enhance the reprogramming efficiency. This study is a step towards developing a functional scaffold through a parallel interpretation of electrospun PLLA scaffolds with two distinct topologies to achieve sustained delivery of two muscle-specific microRNAs (miR-1 and miR-133a) to directly reprogram the adult human cardiac fibroblasts into cardiomyocyte-like cells. Polyethyleneimine was used to form stable PEI-miRNA complexes through electrostatic interactions. These complexes were immobilized on the electrospun smooth and porous scaffolds, where a loading efficiency of ~96% for the fibronectin modified and ~38% for unmodified surfaces was observed, regardless of their surface topology. The in-vitro release experiment exhibited a biphasic release pattern of PEI-miRNA polyplexes from the scaffolds. These dual miRNA scaffold systems proved to be an excellent formulation since their combinatorial effect involving the topographic cues of electrospun fibers, and dual miRNAs helped control the cardiac fibroblast cell fate precisely.
Collapse
Affiliation(s)
- Priyadharshni Muniyandi
- Graduate School of Interdisciplinary New Science, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan
| | - Vivekanandan Palaninathan
- Graduate School of Interdisciplinary New Science, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan; Bio-Nano Electronics Research Centre, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan.
| | - Toru Mizuki
- Graduate School of Interdisciplinary New Science, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan; Bio-Nano Electronics Research Centre, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan
| | - M Sheikh Mohamed
- Graduate School of Interdisciplinary New Science, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan; Bio-Nano Electronics Research Centre, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan
| | - Tatsuro Hanajiri
- Graduate School of Interdisciplinary New Science, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan; Bio-Nano Electronics Research Centre, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan
| | - Toru Maekawa
- Graduate School of Interdisciplinary New Science, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan; Bio-Nano Electronics Research Centre, Toyo University, 2100 Kujirai, Kawagoe, Saitama 3508585, Japan
| |
Collapse
|
42
|
Abu Lila AS, Abdallah MH, Wani SUD, Gangadharappa H, Younes KM, Khafagy ES, Shehata TM, Soliman MS. Folic acid-conjugated raloxifene-loaded graphene-based nanocarrier: Fabrication, characterization and antitumor screening. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.126971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
43
|
Baru A, Sharma S, Purakayastha BPD, Khan S, Mazumdar S, Gupta R, Kundu PK, Arora NM. AXTEX-4D: A Three-Dimensional Ex Vivo Platform for Preclinical Investigations of Immunotherapy Agents. Assay Drug Dev Technol 2021; 19:361-372. [PMID: 34319797 DOI: 10.1089/adt.2021.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The latest advancements in oncology are majorly focused on immuno-oncology (I-O) therapies. However, only ∼7% of drugs are being approved from the preclinical discovery phase to phase 1. The most challenging issues in I-O are the development of active and efficient drugs in an economically feasible way and in a comparatively short time for testing and validation. This mandates an urgent need for the upgradation of preclinical screening models that closely mimic the in vivo tumor microenvironment (TME). The established and most common methods for investigating the tumoricidal activity of I-O drugs are either two-dimensional systems or primary tumor cells in standard tissue culture vessels. Unfortunately, they do not mimic the TME. Consequently, the more in vivo-like three-dimensional (3D) multicellular tumor spheroids are quickly becoming the favored model to examine immune cell-mediated responses in reaction to the administration of I-O drugs. Despite many advantages of multicellular spheroids, challenges (e.g., incompatibility of quantitative assays with spheroid platforms) are still involved in the tedious procedures required for the spheroid culture that is holding back the biological community from adapting the well-recognized spheroid tissue models for studying drug delivery more widely. To this end, we have demonstrated the utility of the 3D ex vivo oncology model, developed on our novel AXTEX-4D™ platform to assess therapeutic efficacies of I-O drugs by investigating immune cell proliferation, migration, infiltration, cytokine profiling, and cytotoxicity of tumor tissueoids. The platform eliminates the need for additional biomolecules such as hydrogels and instead relies on the cancer cells themselves to create their own gradients and microenvironmental factors. In effect, the more comprehensive and ex vivo-like immune-oncology model developed on AXTEX-4D platform can be utilized for high-throughput screening of immunotherapeutic drugs.
Collapse
Affiliation(s)
- Ambica Baru
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | - Swati Sharma
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | | | - Sameena Khan
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | - Saumyabrata Mazumdar
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | - Reeshu Gupta
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | - Prabuddha K Kundu
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| | - Nupur Mehrotra Arora
- Mammalian Cell Culture Lab, Premas Biotech Pvt, Ltd., Sector IV, IMT, Manesar, India
| |
Collapse
|
44
|
10-O-(N N-Dimethylaminoethyl)-Ginkgolide B Methane-Sulfonate (XQ-1H) Ameliorates Cerebral Ischemia Via Suppressing Neuronal Apoptosis. J Stroke Cerebrovasc Dis 2021; 30:105987. [PMID: 34273708 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105987] [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: 07/15/2020] [Revised: 05/30/2021] [Accepted: 06/25/2021] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVES The 10-O-(N N-dimethylaminoethyl)-ginkgolide B methane-sulfonate (XQ-1H) is an effective novel drug for the treatment of ischemic cerebrovascular disease derived from Ginkgolide B, a traditional Chinese medicine, has been widely used in the treatment of cardiovascular and cerebrovascular diseases. However, whether XQ-1H exerts neuroprotective effect via regulating neuronal apoptosis and the underlying mechanism remain to be elucidated. MATERIALS AND METHODS This study was aimed to investigate the neuroprotective effect of XQ-1H in rats subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) and the oxygen glucose deprivation/reoxygenation (OGD/R) induced neuronal apoptosis on pheochromocytoma (PC-12) cells. RESULTS The results showed that administration of XQ-1H at different dosage (7.8, 15.6, 31.2 mg/kg) reduced the brain infarct and edema, attenuated the neuro-behavioral dysfunction, and improved cell morphology in brain tissue after MCAO/R in rats. Moreover, incubation with XQ-1H (1 µM, 3 µM, 10 µM, 50 µM, 100 µM) could increase the cell viability, and showed no toxic effect to PC-12 cells. XQ-1H at following 1 µM, 10 µM, 100 µM decreased the lactate dehydrogenase (LDH) activity and suppressed the cell apoptosis in PC-12 cells exposed to OGD/R. In addition, XQ-1H treatment could significantly inhibit caspase-3 activation both in vivo and in vitro, reciprocally modulate the expression of apoptosis related proteins, bcl-2, and bax via activating PI3K/Akt signaling pathway. For mechanism verification, LY294002, the inhibitor of PI3K/Akt pathway was introduced the expressions of bcl-2 and phosphorylated Akt were down-regulated, the expression of bax was up-regulated, indicating that XQ-1H could alleviate the cell apoptosis through activating the PI3K/Akt pathway. CONCLUSIONS Our findings demonstrated that XQ-1H treatment could provide a neuroprotective effect against ischemic stroke induced by cerebral ischemia/reperfusion injury in vivo and in vitro through regulating neuronal survival and inhibiting apoptosis. The findings of the study confirmed that XQ-1H could be develop as a potential drug for treatment of cerebral ischemic stroke.
Collapse
|
45
|
Bonifácio-Lopes T, Vilas Boas AA, Coscueta ER, Costa EM, Silva S, Campos D, Teixeira JA, Pintado M. Bioactive extracts from brewer's spent grain. Food Funct 2021; 11:8963-8977. [PMID: 33001088 DOI: 10.1039/d0fo01426e] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this study, antioxidant-rich extracts from brewer's spent grain (BSG) extracted by solid-to-liquid extraction using different solvents water and ethanol and their mixtures at two ratios (80% ethanol : water (v/v) and 60% ethanol : water (v/v)) were characterized. Nutritional composition was evaluated for the extracts and for the solid residues obtained after extraction. Additionally, the extracts were analyzed for the total phenolic content and individual phenolic compounds and related biological properties including antioxidant capacity (ABTS; ORAC and DNA protection), antihypertensive capacity, antibacterial activity and antibiofilm capacity. Safety was also demonstrated through genotoxicity and cytotoxicity tests. The results obtained showed that while all the extracts exhibited high antioxidant capacity (except ethanolic extract), the highest values were obtained for the 60% ethanol : water extract. The identification of phenolic compounds using HPLC showed that catechin and vanillin were the main compounds identified with the highest concentration being obtained for 60% ethanol : water extraction. In the biological activity assays, water and hydroethanolic extracts were multifunctional (antioxidant and antihypertensive capacity, antibacterial and antibiofilm activity), and the 80% ethanol : water presented better results in some assays. All were non-genotoxic, but the cytotoxicity was dependent on the extract concentration, with complete safe application for all up to 1 mg mL-1. Therefore, this study shows the potential of a viable green solvent based and low cost extraction recovery method of bioactive compounds from brewer's spent grain.
Collapse
Affiliation(s)
- Teresa Bonifácio-Lopes
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal. and CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Ana A Vilas Boas
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| | - Ezequiel R Coscueta
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| | - Eduardo M Costa
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| | - Sara Silva
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| | - Débora Campos
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| | - José A Teixeira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Manuela Pintado
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| |
Collapse
|
46
|
Effects of Dual Purinoceptor-dependent Approach on Release of Vascular Endothelial Growth Factor From Human Microvascular Endothelial Cell (HMEC-1) and Endothelial Cell Condition. J Cardiovasc Pharmacol 2021; 76:349-359. [PMID: 32569015 DOI: 10.1097/fjc.0000000000000866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the recent years, the awareness of the role purinergic signaling plays as a therapeutic target has increased considerably. The purinoceptor allows the action of extracellular nucleotides (P2 receptors) and intermediary products of their metabolism, such as adenosine (P1 receptors), regulating pivotal processes occurring in the cardiovascular system. This study focuses on a dual purinoreceptor-dependent approach, based on the activation of adenosine P1 receptors with the simultaneous inhibition of P2Y12 receptors that can be used as novel platelet inhibitors in antithrombotic therapy. Endothelial cells are directly exposed to the drugs circulating in the bloodstream. That is why effects of our concept on human microvascular endothelial cells (HMEC-1) were examined in in vitro studies, such as enzyme-linked immunosorbent assay and scratch assays. In response to adenosine receptor agonists, levels of secreted vascular endothelial growth factor varied. Two of them, 5'-N-ethylcarboxamidoadenosine and MRE0094 remarkably increased vascular endothelial growth factor release. The elevated levels were reduced when used together with the P2Y12 receptor antagonist. Also, rates of wound closure in a scratch assay were significantly reduced in these cases. The results suggest that the proposed treatment does not impair endothelial cell condition. In addition, it is suggested as a collateral benefit, namely solving the problem of excessive activation of endothelial cells during antiplatelet therapy.
Collapse
|
47
|
Gdula AM, Swiatkowska M. A2 A receptor agonists and P2Y 12 receptor antagonists modulate expression of thrombospondin-1 (TSP-1) and its secretion from Human Microvascular Endothelial Cells (HMEC-1). Microvasc Res 2021; 138:104218. [PMID: 34182003 DOI: 10.1016/j.mvr.2021.104218] [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: 12/13/2020] [Revised: 05/07/2021] [Accepted: 06/24/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUNDS AND AIMS To address the problem of resistance to standard antiplatelet therapy in some patients, our team proposed a purinoceptor-dependent dual therapy. Its efficacy is also determined by the condition of the vascular endothelium which, by secreting numerous factors, is involved in hemostasis. Among them, thrombospondin-1 is important in the context of thrombotic events. Therefore we sought to determine if the novel dual purinoceptor-dependent concept is associated with TSP-1 changes in vascular endothelial cells. METHODS AND RESULTS TSP-1 expression in human microvascular endothelial cells was determined at transcriptional and protein level. We performed real-time PCR, the Western blot analysis and ELISA test. We found that TSP-1 mRNA and protein expression levels significantly changed in response to P1R agonists treatment. Furthermore, we have observed that co-administration of selective A2AR agonists (UK-432,097 or MRE0094) with P2Y12R antagonists altered TSP-1 expression levels, and the direction of these changes was not synergistic. MRE0094 applied with ARC69931MX or R-138727 increased mRNA expression from 39 to 56 or 57%, respectively (*P < 0.05 vs. MRE0094; ***P < 0.001 vs. control). Also, in the case of the P2Y12R antagonists used together with UK-432,097, there was an increase from 53 to 71 and 70% (*P < 0.05 vs. UK-432,097; ***P < 0.001 vs. control). The observed trends in gene expression were reflected in the protein expression and the level of its secretion from HMEC-1. CONCLUSION The article presents evidence which proves that the purinoceptor-dependent concept is associated with TSP-1 changes in endothelial cells (EC). Moreover, Human Microvascular Endothelial Cells treatment applied together with agonists (MRE0094 or UK-432,097) and P2Y12R antagonist did not result in any synergistic effect, implicating a possible crosstalk between G proteins in GPCRs dependent signaling. Therefore, we suggest that understanding of the specific mechanism underlying TSP-1 alterations in EC in the context of the dual purinoceptor-dependent approach is essential for antiplatelet therapies and should be the subject of future research.
Collapse
Affiliation(s)
- Anna M Gdula
- Department of Cytobiology and Proteomics, Medical University of Lodz, 6/8 Mazowiecka St., 92-215 Lodz, Poland.
| | - Maria Swiatkowska
- Department of Cytobiology and Proteomics, Medical University of Lodz, 6/8 Mazowiecka St., 92-215 Lodz, Poland
| |
Collapse
|
48
|
Evaluation of probiotics for inhibiting hyperproliferation and inflammation relevant to psoriasis in vitro. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
49
|
Kobori T, Tameishi M, Tanaka C, Urashima Y, Obata T. Subcellular distribution of ezrin/radixin/moesin and their roles in the cell surface localization and transport function of P-glycoprotein in human colon adenocarcinoma LS180 cells. PLoS One 2021; 16:e0250889. [PMID: 33974673 PMCID: PMC8112653 DOI: 10.1371/journal.pone.0250889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/15/2021] [Indexed: 12/20/2022] Open
Abstract
The ezrin/radixin/moesin (ERM) family proteins act as linkers between the actin cytoskeleton and P-glycoprotein (P-gp) and regulate the plasma membrane localization and functionality of the latter in various cancer cells. Notably, P-gp overexpression in the plasma membrane of cancer cells is a principal factor responsible for multidrug resistance and drug-induced mutagenesis. However, it remains unknown whether the ERM proteins contribute to the plasma membrane localization and transport function of P-gp in human colorectal cancer cells in which the subcellular localization of ERM has yet to be determined. This study aimed to determine the gene expression patterns and subcellular localization of ERM and P-gp and investigate the role of ERM proteins in the plasma membrane localization and transport function of P-gp using the human colon adenocarcinoma cell line LS180. Using real-time reverse transcription polymerase chain reaction and immunofluorescence analyses, we showed higher levels of ezrin and moesin mRNAs than those of radixin mRNA in these cells and preferential distribution of all three ERM proteins on the plasma membrane. The ERM proteins were highly colocalized with P-gp. Additionally, we show that the knockdown of ezrin, but not of radixin and moesin, by RNA interference significantly decreased the cell surface expression of P-gp in LS180 cells without affecting the mRNA expression of P-gp. Furthermore, gene silencing of ezrin substantially increased the intracellular accumulation of rhodamine123, a typical P-gp substrate, with no alterations in the plasma membrane permeability of Evans blue, a passive transport marker. In conclusion, ezrin may primarily regulate the cell surface localization and transport function of P-gp as a scaffold protein without influencing the transcriptional activity of P-gp in LS180 cells. These findings should be relevant for treating colorectal cancer, which is the second leading cause of cancer-related deaths in males and females combined.
Collapse
Affiliation(s)
- Takuro Kobori
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Mayuka Tameishi
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Chihiro Tanaka
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Yoko Urashima
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Tokio Obata
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
- * E-mail:
| |
Collapse
|
50
|
Cytotoxic Activity of Ethyl-para-methoxycinnamate from Kaempferia galanga L. on A549 Lung Cancer and B16 Melanoma Cancer Cells. JURNAL KIMIA SAINS DAN APLIKASI 2021. [DOI: 10.14710/jksa.24.1.22-28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Kaempferia galanga L. belongs to the family of Zingiberaceae, an endangered medicinal plant with pharmacology activities. Ethyl-p-methoxycinnamate (EPMC) is an essential phytoconstituent of K. galanga rhizomes. Several studies have reported that EPMC has anticancer activities in several cancer cells, including CL-6 gallbladder cancer cells, HepG2liver cancer cells, MCF-7 breast cancer cells, and Raji lymphoma cancer cells. However, studies on A549 lung cancer and B16 melanoma cancer cells have not been reported. This study aimed to determine the anticancer activity of EPMC against A549 lung cancer and B16 melanoma cancer cells. EPMC was obtained by extraction using n-hexane, then recrystallized with chloroform. The isolate was then analyzed by thin-layer chromatography (TLC), and the structure was characterized by Fourier Transform Infrared (FTIR) and Nuclear Magnetic Resonance (NMR) spectroscopy. Cytotoxic activity was determined under Presto Blue assay. Based on the result, EPMC from K. galanga showed the cytotoxic effect on B16 cells with an IC50 value of 97.09 μg/mL, whereas EPMC showed no significant cytotoxic effect on A549 with an IC50 value of 1407.75 μg/mL. It was concluded that EPMC has potential cytotoxic on B16 melanoma cancer cells, but it showed inactive activity against A549 lung cancer cells. Further molecular mechanism underlying EPMC cytotoxic activity needs to be conducted.
Collapse
|