151
|
Morpho-Functional Effect of a New Collagen-Based Medical Device on Human Gingival Fibroblasts: An In Vitro Study. Biomedicines 2023; 11:biomedicines11030786. [PMID: 36979765 PMCID: PMC10045070 DOI: 10.3390/biomedicines11030786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Maintaining periodontal and peri-implant soft tissues health is crucial for the long-term health of teeth and dental implants. New biomedical strategies aimed at avoiding connective tissue alterations and related diseases (e.g., periodontitis and peri-implantitis) are constantly evolving. Among these, collagen-based medical products have proven to be safe and effective. Accordingly, the aim of the present study was to evaluate the effects of Dental SKIN BioRegulation (Guna S.p.a., Milan, Italy), a new injectable medical device composed of type I collagen of porcine origin, on primary cultures of human gingival fibroblasts (hGF). To this end, hGF were cultured on collagen-coated (COL, 100 µg/2 mL) or uncoated plates (CTRL) before evaluating cell viability (24 h, 48 h, 72 h, and 7 d), wound healing properties (3 h, 6 h, 12 h, 24 h, and 48 h), and the activation of mechanotransduction markers, such as FAK, YAP, and TAZ (48 h). The results proved a significant increase in cell viability at 48 h (p < 0.05) and wound closure at 24 h (p < 0.001) of hGF grown on COL, with an increasing trend at all time-points. Furthermore, COL significantly induced the expression of FAK and YAP/TAZ (p < 0.05), thereby promoting the activation of mechanotransduction signaling pathways. Overall, these data suggest that COL, acting as a mechanical bio-scaffold, could represent a useful treatment for gingival rejuvenation and may possibly help in the resolution of oral pathologies.
Collapse
|
152
|
de Dios Andres P, Städler B. Micromotor-Assisted Keratinocytes Migration in a Floating Paper Chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2201251. [PMID: 35694770 DOI: 10.1002/smll.202201251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/21/2022] [Indexed: 06/15/2023]
Abstract
In vitro epidermis models are important to evaluate and study disease progression and possible dermal drug delivery. An in vitro epidermis model using floating paper chips as a scaffold for proliferation and differentiation of primary human keratinocytes is reported. The formation of the four main layers of the epidermis (i.e., basal, spinosum, granulose, and cornified layers) is confirmed. The development of a cornified layer and the tight junction formation are evaluated as well as the alterations of organelles during the differentiation process. Further, this in vitro model is used to assess keratinocyte migration. Finally, magnetic micromotors are assembled, and their ability to aid cell migration on paper chips is confirmed when a static magnetic field is present. Taken together, this attempt to combine bottom-up synthetic biology with dermatology offers interesting opportunities for studying skin disease pathologies and evaluate possible treatments.
Collapse
Affiliation(s)
- Paula de Dios Andres
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| | - Brigitte Städler
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus, 8000, Denmark
| |
Collapse
|
153
|
Bawadud RS, Alkhatib MH. Growth and invasion inhibition of T47D ductal carcinoma cells by the association of docetaxel with a bioactive agent in neutral nanosuspension. BIOIMPACTS : BI 2023; 13:145-157. [PMID: 37193079 PMCID: PMC10182446 DOI: 10.34172/bi.2023.23515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 08/16/2022] [Accepted: 09/13/2022] [Indexed: 05/18/2023]
Abstract
Introduction: The approach for drug delivery has impressively developed with the emergence of nanosuspension, particularly the targeted nanoemulsions (NEs). It can potentially improve the bioavailability of drugs, enhancing their therapeutic efficiency. This study aims to examine the potential role of NE as a delivery system for the combination of docetaxel (DTX), a microtubule-targeting agent, and thymoquinone (TQ) in the treatment of human ductal carcinoma cells T47D. Methods: NEs were synthesized by ultra-sonication and characterized physically by dynamic light scattering (DLS). A sulforhodamine B assay was performed to evaluate cytotoxicity, and a flow cytometry analysis for cell cycle, apoptosis, autophagy, and cancer stem cell evaluations. A quantitative polymerase chain reaction further assessed the epithelial-mesenchymal transition gene expirations of SNAIL-1, ZEB-1, and TWIST-1. Results: The optimal sizes of blank-NEs and NE-DTX+TQ were found at 117.3 ± 8 nm and 373 ± 6.8 nm, respectively. The synergistic effect of the NE-DTX+TQ formulation significantly inhibited the in vitro proliferation of T47D cells. It caused a significant increase in apoptosis, accompanied by the stimulation of autophagy. Moreover, this formulation arrested T47D cells at the G2/M phase, promoted the reduction of the breast cancer stem cell (BCSC) population, and repressed the expression of TWIST-1 and ZEB-1. Conclusion: Co-delivery of NE-DTX+TQ may probably inhibit the proliferation of T47D via the induction of apoptosis and autophagy pathways and impede the migration by reducing the BCSC population and downregulating TWIST-1 expression to decrease the epithelial-to-mesenchymal transition (EMT) of breast cancer cells. Therefore, the study suggests the NE-DTX+TQ formula as a potential approach to inhibit breast cancer growth and metastasis.
Collapse
Affiliation(s)
- Raghdah S. Bawadud
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mayson H. Alkhatib
- Department of Biological Sciences & Chemistry, College of Arts and Sciences, University of Nizwa, Nizwa, Sultanate of Oman
| |
Collapse
|
154
|
Zhang T, Song C, Zheng C, Chen X, Zhang Y. Extracellular Amyloid β-protein (1-42) Oligomers Anchor Brain Cells and Make them inert as an Unconventional Integrin-Coupled Ligand. Cell Mol Neurobiol 2023; 43:841-858. [PMID: 35445880 PMCID: PMC11415169 DOI: 10.1007/s10571-022-01219-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/30/2022] [Indexed: 12/12/2022]
Abstract
This study aimed to investigate the effect of extracellular Aβ42 on neural cell migration, and the possible molecular mechanisms. Extracellular Aβ42 monomers did not negatively affect the motility of neural cells; however, they could promote cell migration from toxic extracellular Aβ42 oligomers. Contrastingly, extracellular Aβ42 aggregates, especially Aβ42 oligomers, significantly decreased neural cell migration while reducing their survival. Further, their soluble and deposited states showed different effects in causing the neural cells to become inert (incapable of moving). These findings were consistent with that of binding of Aβ42 oligomers to the plasma membrane or integrin receptors of the inert cells. By combining the protection of cell migratory capability by anti-oligomeric Aβ42 scFv antibody with the information obtained from our docking model of the Aβ42 trimer and integrin molecule, our findings suggest that extracellular Aβ42 aggregates disrupt the function of integrins mainly through the RHDS motif of Aβ42 chain, which eventually causes neural cells to become inert. Thus, we propose an "anchor" opinion, where Aβ42 aggregates in the ECM serve as the adverse "anchors" in the brain for anchoring neurons and for making neural cells inert, which causes their dysfunction. The neural cells with damaged motility could be restored or repaired if these anchoring effects of extracellular Aβ42 aggregates on the neural cells were severed or reduced, even if the "anchors" themselves were not completely eliminated. Medicines targeting soluble and deposited anchors of Aβ42 aggregates could be developed into effective treatments for Alzheimer disease.
Collapse
Affiliation(s)
- Tianyu Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun, 130012, China
| | - Chuli Song
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun, 130012, China
| | - Changxin Zheng
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun, 130012, China
| | - Xu Chen
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun, 130012, China
| | - Yingjiu Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun, 130012, China.
- School of Life Science, Jilin University, Changchun, 130012, China.
| |
Collapse
|
155
|
Bu S, Joseph JJ, Nguyen HC, Ehsan M, Rasheed B, Singh A, Qadura M, Frisbee JC, Singh KK. MicroRNA miR-378-3p is a novel regulator of endothelial autophagy and function. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 3:100027. [PMID: 39803361 PMCID: PMC11708318 DOI: 10.1016/j.jmccpl.2022.100027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 01/16/2025]
Abstract
Autophagy is a highly conserved cellular process in which cytoplasmic materials are internalized into an autophagosome that later fuses with a lysosome for their degradation and recycling. MicroRNAs (miRNAs) are integral regulators in various cellular processes including autophagy and endothelial function. Accordingly, we hypothesize that miRNA, miR-378-3p, is an essential regulator of endothelial autophagy and endothelial function. MiR-378-3p expression was measured following inhibition and activation of autophagy in endothelial cells. A gain- or loss-of function approach was employed to either overexpress or inhibit the expression of miR-378-3p, respectively, in cultured endothelial cells, and markers of autophagy and indices of endothelial function, such as proliferation, migration and tube forming potential were measured. Inhibition and activation of autophagy up- and down-regulated the expression of miR-378-3p, respectively. Furthermore, miR-378a-3p overexpression was associated with impaired autophagy indicated by a reduced LC3-II/LC3-I ratio, and endothelial function indicated by increased proliferation associated with reduced p21 expression, reduced angiogenic potential and increased migration, which were associated with reduced expression of endothelial nitric oxide synthase (eNOS), an essential regulator of endothelial function. Accordingly, miR-378a-3p inhibition was associated with reduced cell proliferation, migration and increased eNOS in endothelial cells. Apoptosis was not affected in cells transfected with antagomir. Using in silico approach, Protein Disulfide Isomerase Family A Member 4 (PDIA-4) was identified and confirmed as a target of miR-378-3p. PDIA-4 expression was significantly reduced or enhanced in miR-378-3p-overexpressing or -silenced endothelial cells, respectively. Our findings show an inverse relationship between miR-378-3p and endothelial autophagy and function, providing a novel insight about the epigenetic regulation of these processes.
Collapse
Affiliation(s)
- Shuhan Bu
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Jameela J. Joseph
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Hien C. Nguyen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Mehroz Ehsan
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Berk Rasheed
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Aman Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Mohammad Qadura
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Jefferson C. Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Krishna K. Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
156
|
Ramzan F, Nabi SA, Lone MS, Imtiyaz K, Urooj L, Vishakha V, Sharma K, Rizvi MMA, Shafi S, Samim M, Bano S, Javed K. Synthesis, Molecular Docking, and Biological Evaluation of a New Series of Benzothiazinones and Their Benzothiazinyl Acetate Derivatives as Anticancer Agents against MCF-7 Human Breast Cancer Cells and as Anti-Inflammatory Agents. ACS OMEGA 2023; 8:6650-6662. [PMID: 36844525 PMCID: PMC9948161 DOI: 10.1021/acsomega.2c07153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
Six 1,4-benzothiazin-3-ones (2a-f) and four benzothiazinyl acetate derivatives (3a-d) were synthesized and characterized by various spectroscopic methods, namely, 1H NMR, 13C NMR, IR, MS, and elemental analysis. The cytotoxic effects of the compounds were assessed against MCF-7, a human breast cancer cell line, along with their anti-inflammatory activity. Molecular docking studies performed against the VEGFR2 kinase receptor displayed a common binding orientation of the compounds in the catalytic binding pocket of the receptor. The generalized Born surface area (GBSA) studies of compound 2c with the highest docking score also proved its stability in binding to the kinase receptor. Compounds 2c and 2b showed better results against VEGFR2 kinase with IC50 values of 0.0528 and 0.0593 μM, respectively, compared to sorafenib. All of the compounds (2a-f and 3a-d) showed effective growth inhibition having (IC50) values of 2.26, 1.37, 1.29, 2.30, 4.98, 3.7, 5.19, 4.50, 4.39, and 3.31 μM, respectively, against the MCF-7 cell line compared to standard 5-fluorouracil (IC50 = 7.79 μM). However, compound 2c displayed remarkable cytotoxic activity (IC50 = 1.29 μM), suggesting it as a lead compound in the cytotoxic assay. Additionally, compounds 2c and 2b showed better results against VEGFR2 kinase with IC50 values of 0.0528 and 0.0593 μM, respectively, compared to sorafenib. It also inhibited hemolysis by stabilizing the membrane comparable to that of diclofenac sodium, a standard used in the human red blood cell membrane stabilization assay and hence can act as a template for designing novel anticancer and anti-inflammatory agents.
Collapse
Affiliation(s)
- Farhat Ramzan
- Department
of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Syed Ayaz Nabi
- Department
of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Mehak Saba Lone
- Department
of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Khalid Imtiyaz
- Department
of Biosciences, Genome biology lab, Jamia
Millia Islamia, New Delhi 110025, India
| | - Laraib Urooj
- Department
of Biosciences, Genome biology lab, Jamia
Millia Islamia, New Delhi 110025, India
| | - Vishakha Vishakha
- Central
European Institute of Technology, Brno University
of Technology, Brno 61200, Czech Republic
| | - Kalicharan Sharma
- Department
of Pharmaceutical Chemistry, Delhi Pharmaceutical
Sciences and Research University Pushpvihar, New Delhi 110017, India
| | - M. Moshahid A. Rizvi
- Department
of Biosciences, Genome biology lab, Jamia
Millia Islamia, New Delhi 110025, India
| | - Syed Shafi
- Department
of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Mohammed Samim
- Department
of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Sameena Bano
- Department
of Computer Science and Engineering, School of Engineering Sciences
and Technology, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Kalim Javed
- Department
of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| |
Collapse
|
157
|
Rapid generation of homogenous tumor spheroid microtissues in a scaffold-free platform for high-throughput screening of a novel combination nanomedicine. PLoS One 2023; 18:e0282064. [PMID: 36800370 PMCID: PMC9937506 DOI: 10.1371/journal.pone.0282064] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Combination nanomedicine is a potent strategy for cancer treatment. Exploiting different mechanisms of action, a novel triple drug delivery system of 5-fluorouracil, curcumin, and piperine co-loaded human serum albumin nanoparticles (5FU-CUR-PIP-HSA-NPs) was developed via the self-assembly method for suppressing breast tumor. Both hydrophobic and hydrophilic drugs were successfully encapsulated in the HSA NPs with a high drug loading efficiency (DLE) of 10%. Successful clinical translation of nanomedicines, however, is a challenging process requiring considerable preclinical in vitro and in vivo animal tests. The aim of this study was to develop a homemade preclinical 3D culture model in the standard 96-well plates in a cost and time-effective novel approach for the rapid generation of homogenous compact tumor spheroids for disease modeling, and anticancer therapeutic/nanomedicine screening. The knowledge of drug screening can be enhanced by employing such a model in a high-throughput manner. Accordingly, to validate the formulated drug delivery system and investigate the cellular uptake and cytotoxicity effect of the nanoformulation, 3D tumor spheroids were employed. The practicality of the nanomedicine system was substantiated in different tests. The in vitro uptake of the NPs into the tight 3D tumor spheroids was facilitated by the semi-spherical shape of the NPs with a proper size and surface charge. 5FU-CUR-PIP-HSA-NPs demonstrated high potency of migration inhibition as a part of successful anti-metastatic therapy as well. The remarkable differences in 2D and 3D cytotoxicities emphasize the importance of employing 3D tumor models as an intermediate step prior to in vivo animal experiments for drug/nanomedicine screening.
Collapse
|
158
|
Shelley JR, McHugh BJ, Wills J, Dorin JR, Weller R, Clarke DJ, Davidson DJ. A mechanistic evaluation of human beta defensin 2 mediated protection of human skin barrier in vitro. Sci Rep 2023; 13:2271. [PMID: 36755116 PMCID: PMC9908873 DOI: 10.1038/s41598-023-29558-0] [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: 11/07/2022] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
The human skin barrier, a biological imperative, is impaired in inflammatory skin diseases such as atopic dermatitis (AD). Staphylococcus aureus is associated with AD lesions and contributes to pathological inflammation and further barrier impairment. S. aureus secretes extracellular proteases, such as V8 (or 'SspA'), which cleave extracellular proteins to reduce skin barrier. Previous studies demonstrated that the host defence peptide human beta-defensin 2 (HBD2) prevented V8-mediated damage. Here, the mechanism of HBD2-mediated barrier protection in vitro is examined. Application of exogenous HBD2 provided protection against V8, irrespective of timeline of application or native peptide folding, raising the prospect of simple peptide analogues as therapeutics. HBD2 treatment, in context of V8-mediated damage, modulated the proteomic/secretomic profiles of HaCaT cells, altering levels of specific extracellular matrix proteins, potentially recovering V8 damage. However, HBD2 alone did not substantially modulate cellular proteomic/secretomics profiles in the absence of damage, suggesting possible therapeutic targeting of lesion damage sites only. HBD2 did not show any direct protease inhibition or induce expression of known antiproteases, did not alter keratinocyte migration or proliferation, or form protective nanonet structures. These data validate the barrier-protective properties of HBD2 in vitro and establish key protein datasets for further targeted mechanistic analyses.
Collapse
Affiliation(s)
- Jennifer R Shelley
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, BioQuarter, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK.
- The Commonwealth Building, The Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
| | - Brian J McHugh
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, BioQuarter, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Jimi Wills
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital Campus, Crewe Road, Edinburgh, EH4 2XU, Scotland, UK
| | - Julia R Dorin
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, BioQuarter, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Richard Weller
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, BioQuarter, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - David J Clarke
- The EastChem School of Chemistry, University of Edinburgh, Joseph Black Building, Brewster Road, Edinburgh, EH9 3FJ, UK
| | - Donald J Davidson
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, BioQuarter, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| |
Collapse
|
159
|
Mclaughlin M, Hesketh KL, Horgan SL, Florida-James G, Cocks M, Strauss JA, Ross M. Ex Vivo treatment of coronary artery endothelial cells with serum post-exercise training offers limited protection against in vitro exposure to FEC-T chemotherapy. Front Physiol 2023; 14:1079983. [PMID: 36818448 PMCID: PMC9932712 DOI: 10.3389/fphys.2023.1079983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023] Open
Abstract
Background: Chemotherapy treatment for breast cancer associates with well-documented cardiovascular detriments. Exercise has shown promise as a potentially protective intervention against cardiac toxicity. However, there is a paucity of evidence for the benefits of exercise on the vasculature. Objectives: This study aimed to determine the effects of chemotherapy on the vascular endothelium; and if there are protective effects of serological alterations elicited by an exercise training intervention. Methods and Results: 15 women participated in a 12-week home-based exercise intervention consisting of three high-intensity interval sessions per week. Human coronary artery endothelial cells (HCAEC) were exposed to physiological concentrations of 5-fluorouracil, epirubicin, cyclophosphamide (FEC) and docetaxel to determine a dose-response. Twenty-4 hours prior to FEC and docetaxel exposure, HCAECs were preconditioned with serum collected pre- and post-training. Annexin V binding and cleaved caspase-3 were assessed using flow cytometry and wound repair by scratch assays. Chemotherapy exposure increased HCAEC Annexin V binding, cleaved caspase-3 expression in a dose-dependent manner; and inhibited wound repair. Compared to pre-training serum, conditioning HCAECs with post-training serum, reduced Annexin V binding (42% vs. 30%, p = 0.01) when exposed to FEC. For docetaxel, there were no within-group differences (pre-vs post-exercise) for Annexin V binding or cleaved caspase-3 expression. There was a protective effect of post-training serum on wound repair for 5-flurouracil (p = 0.03) only. Conclusion: FEC-T chemotherapy drugs cause significant damage and dysfunction of endothelial cells. Preconditioning with serum collected after an exercise training intervention, elicited some protection against the usual toxicity of FEC-T, when compared to control serum.
Collapse
Affiliation(s)
- Marie Mclaughlin
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom,School of Health and Life Sciences, University of the West of Scotland, Lanarkshire, United Kingdom
| | - Katie L. Hesketh
- Liverpool John Moores University, School of Sport and Exercise Sciences, Liverpool, United Kingdom
| | - Sarah L. Horgan
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| | | | - Matthew Cocks
- Liverpool John Moores University, School of Sport and Exercise Sciences, Liverpool, United Kingdom
| | - Juliette A. Strauss
- Liverpool John Moores University, School of Sport and Exercise Sciences, Liverpool, United Kingdom
| | - Mark Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom,School of Energy, Geoscience, Infrastructure and Society, Heriot Watt University, Edinburgh, United Kingdom,*Correspondence: Mark Ross,
| |
Collapse
|
160
|
Ghannam IAY, El Kerdawy AM, Mounier MM, Abo-Elfadl MT, Ali IH. Novel 2-oxo-2-phenylethoxy and benzyloxy diaryl urea hybrids as VEGFR-2 inhibitors: Design, synthesis, and anticancer evaluation. Arch Pharm (Weinheim) 2023; 356:e2200341. [PMID: 36398495 DOI: 10.1002/ardp.202200341] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/11/2022] [Accepted: 10/07/2022] [Indexed: 11/19/2022]
Abstract
Two series of diaryl urea derivatives, 6a-k and 7a-n, were synthesized. All the newly synthesized compounds were tested against the NCI (US) cancer cell lines via SRB assay. The p-chloro-m-trifluoromethyl phenyl derivatives 6e-g and 7e-g showed the most potent cytotoxic activity with a GI50 value range of 1.2-15.9 µM. Furthermore, the p-fluorobenzyloxy diaryl urea derivative 7g revealed the most potent cytotoxicity against eight cancer cell lines in the MTT assay with IC50 values below 5 µM. Compounds 6a-k and 7a-n were tested for their vascular endothelial growth factor receptor-2 (VEGFR-2) kinase inhibitory activities. The p-chloro-m-trifluoromethyl diaryl urea benzyloxy derivatives 7e-i and the p-methoxydiaryl urea benzyloxy derivatives 7k, 7l, and 7n were found to be the most active compounds as VEGFR-2 inhibitors in the benzyloxy series 7, with an IC50 range of 0.09-4.15 µM. In the 2-oxo-2-phenylethoxy series 6, compounds 6e-g and 6i were reported with IC50 values of 0.94, 0.54, 2.71, and 4.81 µM, respectively. Moreover, compounds 7e and 7g induced apoptosis, causing cell cycle arrest in the G2/M phase. In addition, 7g showed an antimigratory effect in A-375 cells and inhibited the VEGFR-2 expression in an immunohistofluorescence study. Molecular docking simulations on VEGFR-2 as well as ADME properties prediction were also performed.
Collapse
Affiliation(s)
- Iman A Y Ghannam
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Egypt
| | - Ahmed M El Kerdawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt.,Department of Pharmaceutical Chemistry, School of Pharmacy, Newgiza University (NGU), Newgiza, Egypt
| | - Marwa M Mounier
- Department of Pharmacognosy, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Egypt
| | - Mahmoud T Abo-Elfadl
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, Egypt.,Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki, Egypt
| | - Islam H Ali
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Egypt
| |
Collapse
|
161
|
Xu N, Tian H, Po Fung C, Lin Y, Chen Y, Zhu G, Shen Y, Guo C, Yang H. Inhibition of human oral squamous cell carcinoma proliferation and migration by prodrug-activating suicide gene therapies. Exp Ther Med 2023; 25:92. [PMID: 36761002 PMCID: PMC9905654 DOI: 10.3892/etm.2023.11790] [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: 06/12/2022] [Accepted: 11/09/2022] [Indexed: 01/11/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), which originates from mucosal epithelium in the oral cavity, pharynx and larynx, is the sixth most common malignancy in the world. The prognosis of HNSCC is not satisfactory due to metastasis, resulting in 5-year survival rates ranging from 65.9 to 67.2%. Previously, we developed a method to evaluate the effect prodrug-activating suicide gene (PA-SG) therapy on the proliferation of HNSCC. The present study investigated PA-SG therapy on metastatic HNSCC by wound-healing assay and our previously established method. HSC-3 cells with stable expression of suicide genes thymidine kinase (TK) or cytosine deaminase (CD) were treated with prodrugs ganciclovir (GCV) or 5-fluorocytosine (5-FC), respectively. Both GCV and 5-FC inhibited HSC-3 proliferation while the bystander effect of CD/5-FC was greater compared with that of TK/GCV. GCV showed a greater anti-migration effect compared with that of 5-FC. To the best of our knowledge, the present study is the first to evaluate the anti-migratory and anti-proliferative effects of PA-SG therapies on metastatic HNSCC. This may also serve as a general method to quantify other types of PA-SC therapy. The present results demonstrated that PA-SG therapy is a promising treatment for anti-metastatic HNSCC therapy development.
Collapse
Affiliation(s)
- Naining Xu
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518001, P.R. China
- Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, Guangdong 518001, P.R. China
| | - Honglei Tian
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong 999077, P.R. China
| | - Chun Po Fung
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong 999077, P.R. China
| | - Yuntao Lin
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518001, P.R. China
- Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, Guangdong 518001, P.R. China
| | - Yuling Chen
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518001, P.R. China
- Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, Guangdong 518001, P.R. China
| | - Guang Zhu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong 999077, P.R. China
| | - Yuehong Shen
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518001, P.R. China
- Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, Guangdong 518001, P.R. China
| | - Chuanbin Guo
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| | - Hongyu Yang
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518001, P.R. China
- Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Shenzhen, Guangdong 518001, P.R. China
| |
Collapse
|
162
|
Solbu AA, Caballero D, Damigos S, Kundu SC, Reis RL, Halaas Ø, Chahal AS, Strand BL. Assessing cell migration in hydrogels: An overview of relevant materials and methods. Mater Today Bio 2023; 18:100537. [PMID: 36659998 PMCID: PMC9842866 DOI: 10.1016/j.mtbio.2022.100537] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/05/2022] [Accepted: 12/28/2022] [Indexed: 12/29/2022] Open
Abstract
Cell migration is essential in numerous living processes, including embryonic development, wound healing, immune responses, and cancer metastasis. From individual cells to collectively migrating epithelial sheets, the locomotion of cells is tightly regulated by multiple structural, chemical, and biological factors. However, the high complexity of this process limits the understanding of the influence of each factor. Recent advances in materials science, tissue engineering, and microtechnology have expanded the toolbox and allowed the development of biomimetic in vitro assays to investigate the mechanisms of cell migration. Particularly, three-dimensional (3D) hydrogels have demonstrated a superior ability to mimic the extracellular environment. They are therefore well suited to studying cell migration in a physiologically relevant and more straightforward manner than in vivo approaches. A myriad of synthetic and naturally derived hydrogels with heterogeneous characteristics and functional properties have been reported. The extensive portfolio of available hydrogels with different mechanical and biological properties can trigger distinct biological responses in cells affecting their locomotion dynamics in 3D. Herein, we describe the most relevant hydrogels and their associated physico-chemical characteristics typically employed to study cell migration, including established cell migration assays and tracking methods. We aim to give the reader insight into existing literature and practical details necessary for performing cell migration studies in 3D environments.
Collapse
Affiliation(s)
- Anita Akbarzadeh Solbu
- Department of Biotechnology and Food Sciences, NOBIPOL, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
| | - David Caballero
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal
| | - Spyridon Damigos
- Department of Biotechnology and Food Sciences, NOBIPOL, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
| | - Subhas C. Kundu
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal
| | - Øyvind Halaas
- Department of Clinical and Molecular Medicine, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
| | - Aman S. Chahal
- Department of Biotechnology and Food Sciences, NOBIPOL, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Berit L. Strand
- Department of Biotechnology and Food Sciences, NOBIPOL, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
163
|
Nguyen HC, Bu S, Nikfarjam S, Rasheed B, Michels DCR, Singh A, Singh S, Marszal C, McGuire JJ, Feng Q, Frisbee JC, Qadura M, Singh KK. Loss of fatty acid binding protein 3 ameliorates lipopolysaccharide-induced inflammation and endothelial dysfunction. J Biol Chem 2023; 299:102921. [PMID: 36681124 PMCID: PMC9988587 DOI: 10.1016/j.jbc.2023.102921] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 01/20/2023] Open
Abstract
Circulating fatty acid-binding protein 3 (FABP3) is an effective biomarker of myocardial injury and peripheral artery disease (PAD). The endothelium, which forms the inner most layer of every blood vessel, is exposed to higher levels of FABP3 in PAD or following myocardial injury, but the pathophysiological role of endothelial FABP3, the effect of FABP3 exposure on endothelial cells, and related mechanisms are unknown. Here, we aimed to evaluate the pathophysiological role of endothelial FABP3 and related mechanisms in vitro. Our molecular and functional in vitro analyses show that (1) FABP3 is basally expressed in endothelial cells; (2) inflammatory stress in the form of lipopolysaccharide (LPS) upregulated endothelial FABP3 expression; (3) loss of endogenous FABP3 protected endothelial cells against LPS-induced endothelial dysfunction; however, exogenous FABP3 exposure exacerbated LPS-induced inflammation; (4) loss of endogenous FABP3 protected against LPS-induced endothelial dysfunction by promoting cell survival and anti-inflammatory and pro-angiogenic signaling pathways. Together, these findings suggest that gain-of endothelial FABP3 exacerbates, whereas loss-of endothelial FABP3 inhibits LPS-induced endothelial dysfunction by promoting cell survival and anti-inflammatory and pro-angiogenic signaling. We propose that an increased circulating FABP3 in myocardial injury or PAD patients may be detrimental to endothelial function, and therefore, therapies aimed at inhibiting FABP3 may improve endothelial function in diseased states.
Collapse
Affiliation(s)
- Hien C Nguyen
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Shuhan Bu
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Sepideh Nikfarjam
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Berk Rasheed
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - David C R Michels
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Aman Singh
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Shweta Singh
- Department of Applied Science, Fanshawe College, London, Ontario, Canada
| | - Caroline Marszal
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - John J McGuire
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Qingping Feng
- Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Jefferson C Frisbee
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mohammad Qadura
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Krishna K Singh
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
164
|
Comparison of in vitro scratch wound assay experimental procedures. Biochem Biophys Rep 2023; 33:101423. [PMID: 36647554 PMCID: PMC9840221 DOI: 10.1016/j.bbrep.2023.101423] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Fibroblast migration is an important aspect of wound healing. Different factors can influence migration and as such proper wound healing. In vitro scratch wound assays are used to examine cellular migration. However, the wide array of techniques available reduces reproducibility of findings. In this paper, we compare two techniques for wound creation; i.e. the exclusion method or scratching of cell monolayers. Furthermore, we investigate if analysis software influences experimental outcome by comparing both commercially and freely available analysis software. Besides, we examine the effect of cortisol on migration behavior of fibroblasts and identify possible caveats in experimental design. Results show a significantly reduced migration of fibroblasts when wounds are created using a cell exclusion method. Furthermore, addition of cortisol to the cell culture media only reduced migration of fibroblast monolayers that had been scratched but not in those where wounds were created using the exclusion method. A possible explanation related to cytokine expression is discussed.
Collapse
|
165
|
Bai Q, Wang X, Yan H, Wen L, Zhou Z, Ye Y, Jing Y, Niu Y, Wang L, Zhang Z, Su J, Chang T, Dou G, Wang Y, Sun J. Microglia-Derived Spp1 Promotes Pathological Retinal Neovascularization via Activating Endothelial Kit/Akt/mTOR Signaling. J Pers Med 2023; 13:jpm13010146. [PMID: 36675807 PMCID: PMC9866717 DOI: 10.3390/jpm13010146] [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: 11/18/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Pathological retinal neovascularization (RNV) is the main character of ischemic ocular diseases, which causes severe visual impairments. Though retinal microglia are well acknowledged to play important roles in both physiological and pathological angiogenesis, the molecular mechanisms by which microglia communicates with endothelial cells (EC) remain unknown. In this study, using single-cell RNA sequencing, we revealed that the pro-inflammatory secreted protein Spp1 was the most upregulated gene in microglia in the mouse model of oxygen-induced retinopathy (OIR). Bioinformatic analysis showed that the expression of Spp1 in microglia was respectively regulated via nuclear factor-kappa B (NF-κB) and hypoxia-inducible factor 1α (HIF-1α) pathways, which was further confirmed through in vitro assays using BV2 microglia cell line. To mimic microglia-EC communication, the bEnd.3 endothelial cell line was cultured with conditional medium (CM) from BV2. We found that adding recombinant Spp1 to bEnd.3 as well as treating with hypoxic BV2 CM significantly enhanced EC proliferation and migration, while Spp1 neutralizing blocked those CM-induced effects. Moreover, RNA sequencing of BV2 CM-treated bEnd.3 revealed a significant downregulation of Kit, one of the type III tyrosine kinase receptors that plays a critical role in cell growth and activation. We further revealed that Spp1 increased phosphorylation and expression level of Akt/mTOR signaling cascade, which might account for its pro-angiogenic effects. Finally, we showed that intravitreal injection of Spp1 neutralizing antibody attenuated pathological RNV and improved visual function. Taken together, our work suggests that Spp1 mediates microglia-EC communication in RNV via activating endothelial Kit/Akt/mTOR signaling and is a potential target to treat ischemic ocular diseases.
Collapse
Affiliation(s)
- Qian Bai
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- 63750 Army Hospital of Chinese PLA, Xi’an 710043, China
| | - Xin Wang
- Lintong Rehabilitation Center of PLA Joint Logistics Support Force, Xi’an 710600, China
| | - Hongxiang Yan
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Lishi Wen
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Ziyi Zhou
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Yating Ye
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- College of Life Science, Northwestern University, Xi’an 710069, China
| | - Yutong Jing
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Yali Niu
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Liang Wang
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Department of Ophthalmology, The Northern Theater Air Force Hospital, Shenyang 110041, China
| | - Zifeng Zhang
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Jingbo Su
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Tianfang Chang
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Guorui Dou
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Yusheng Wang
- Eye Institute of Chinese PLA, Fourth Military Medical University, Xi’an 710032, China
- Correspondence: (Y.W.); (J.S.); Tel.: +029-84775371 (Y.W.); +029-84771273 (J.S.)
| | - Jiaxing Sun
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, China
- Correspondence: (Y.W.); (J.S.); Tel.: +029-84775371 (Y.W.); +029-84771273 (J.S.)
| |
Collapse
|
166
|
Wang J, Ding Y, Li D, Zhu N, Nishiyama A, Yuan Y. (Pro)renin receptor promotes colorectal cancer progression through inhibiting the NEDD4L-mediated Wnt3 ubiquitination and modulating gut microbiota. Cell Commun Signal 2023; 21:2. [PMID: 36597142 PMCID: PMC9809055 DOI: 10.1186/s12964-022-01015-x] [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: 06/05/2022] [Accepted: 12/11/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND We previously found that (pro)renin receptor ((P)RR) augments Wnt3 protein without affecting Wnt3 gene transcription in colorectal cancer (CRC) cells, thus contributes to CRC initiation. The present study aims to investigate whether (P)RR further promotes CRC progression following oncogenesis and the related mechanisms. Notably, we deeply elaborate how (P)RR affects Wnt3 protein level and the key enzyme that mediates this process. METHODS Immunohistochemistry, western blotting and immunofluorescence were performed to detect protein expression status. A kind of gastrointestinal epithelium-specific ATP6AP2 ((P)RR encoding gene) knock-in mice were generated using Crispr/Cas9 system. RESULTS We found that increased (P)RR expression in primary CRC lesions is positively associated with higher Wnt3 protein level and disease progression. Progressive CRC presents less colocalization of Wnt3 and an E3 ubiquitin ligase NEDD4L in primary lesions than non-progressive CRC. In colon cancer cells, (P)RR dramatically inhibits the NEDD4L-mediated Wnt3 protein ubiquitination. ATP6AP2 knock-in mice show more diminished Wnt3-NEDD4L colocalization in their gut epithelium in comparison to wildtype mice. They also have abnormal gut bacterial flora distribution. Especially, Lachnospiraceae_NK4A136 and Bacteroides genus, which are generally protective against CRC, are suppressed in guts of ATP6AP2 knock-in mice. CONCLUSIONS Collectively, (P)RR promotes CRC progression through inhibiting the NEDD4L-mediated Wnt3 ubiquitination and modulating gut microbiota. Video Abstract.
Collapse
Affiliation(s)
- Juan Wang
- grid.412465.0Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences of Zhejiang Province), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China ,Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center of Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Yuwei Ding
- grid.412465.0Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences of Zhejiang Province), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China ,Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center of Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Dan Li
- grid.412465.0Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences of Zhejiang Province), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China ,Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center of Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Ning Zhu
- grid.412465.0Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences of Zhejiang Province), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China ,Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center of Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Akira Nishiyama
- grid.258331.e0000 0000 8662 309XDepartment of Pharmacology, Faculty of Medicine, Kagawa University, Takamatsu, Kagawa 761-0793 Japan
| | - Ying Yuan
- grid.412465.0Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences of Zhejiang Province), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China ,Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center of Zhejiang University, Hangzhou, 310058 Zhejiang China
| |
Collapse
|
167
|
Liu J, Li Y, Jiang Z, Liu Y, Wei Z. Protein arginine methyltransferase 1 upregulates matrix metalloproteinase-2/9 expression via Zeste Homolog 2 to promote human rheumatoid arthritis fibroblast-like synovial cell survival and metastasis. Int J Rheum Dis 2023; 26:88-98. [PMID: 36239067 DOI: 10.1111/1756-185x.14454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/28/2022] [Accepted: 09/22/2022] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To explore the role of protein arginine methyltransferase 1 (PRMT1) in the development of rheumatoid arthritis (RA). METHODS Fibroblast-like synoviocytes (FLS) were isolated from synovial tissues, cultured and transfected with plasmid vector or short hairpin RNA (shRNA). The morphology and surface markers of FLS were investigated by light microscopy and flow cytometry. The expression levels of PRMT1, Zeste Homolog 2 (EZH2), matrix metalloproteinase-2 (MMP-2) and MMP-9 were detected by real-time polymerase chain reaction and Western blotting. Cell viability was tested by MTT assay, cell proliferation was assessed by EdU assay, cell invasion was evaluated by Transwell invasion test, cell migration was detected by wound healing assay, and cell apoptosis was detected by flow cytometry. RESULTS The expression of PRMT1 was elevated in RA synovial tissues compared with control tissues. FLS from control tissues showed a lower level of PRMT1 than FLS from RA tissues, and FLS from RA tissues had a stronger ability of cell survival and metastasis than those from control tissues. When silencing PRMT1 expression, FLS from RA tissues showed a decreased ability of cell survival and metastasis. Additionally, FLS from RA tissues expressed a higher level of MMP-2 and MMP-9. When silencing PRMT1 expression, the expression of MMP-2 and MMP-9 of FLS was suppressed. Furthermore, the effect of PRMT1 on FLS was mediated by EZH2. CONCLUSION We found that PRMT1 had an overall effect on FLS via EZH2, which contributed to the development of RA. Hence, PRMT1 and EZH2 provide potential targets for RA treatment.
Collapse
Affiliation(s)
- Juan Liu
- Department of Rheumatology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yongsheng Li
- Department of Rheumatology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Zhen Jiang
- Department of Rheumatology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yan Liu
- Department of Rheumatology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Zhongcheng Wei
- Department of Cardiology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| |
Collapse
|
168
|
Yan W, Li Y, Liu Y, Wen Y, Pei H, Yang J, Chen L. Crystal structure of tubulin-barbigerone complex enables rational design of potent anticancer agents with isoflavone skeleton. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154550. [PMID: 36610121 DOI: 10.1016/j.phymed.2022.154550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Isoflavones possess many biological activities, including anti-inflammatory and anticancer effects. Microtubules (composed of αβ-tubulin heterodimers) are described as one possible cellular target of some of these isoflavones. However, the binding of tubulin to isoflavones has not been extensively studied, and until now, no crystal structure of the tubulin-isoflavone complex has been solved, and details of the isoflavone-tubulin interaction remain elusive. PURPOSE Barbigerone is an isoflavone mainly found in the genus Milletti, such as the edible leguminous plant Millettia ferruginea, with anticancer activity. This study aims to confirm the cellular target of barbigerone and to study its anticancer mechanism. METHOD Surface plasmon resonance assays and X-ray crystallography were used to study the interaction of barbigerone with tubulin protein. Immunofluorescence, in-cell and in vitro tubulin polymerization assays were employed to investigate the mechanism. MTT assays, cell clonal formation assays, wound healing assays, tube formation assays and H460 xenograft models were conducted to evaluate the in vitro and in vivo anticancer activities of barbigerone and one of its derivatives, 0412. RESULTS Here, we found that barbigerone binds to tubulin to inhibit tubulin polymerization. Moreover, we solved the X-ray crystal structure of the tubulin-barbigerone complex at 2.33 Å resolution, which unambiguously determined the orientation and position of barbigerone in the colchicine-binding site. Illuminated by the X-ray data, we synthetized and obtained a more active isoflavone, 0412. Both barbigerone and 0412 inhibit cancer cell proliferation, tubulin polymerization, migration of HeLa cells and capillary-like tube formation of HUVECs, induce G2/M phase cell cycle arrest and apoptosis, and exhibit anticancer activity in an H460 xenograft model. CONCLUSION In all, through biochemical and X-ray crystal structure results, we identified tubulin as the cellular target of one isoflavone, barbigerone, and proved that the tubulin-barbigerone complex plays a guiding role in obtaining a more active compound, 0412. These studies provide a crucial research basis for the development of isoflavones as anticancer candidate compounds.
Collapse
Affiliation(s)
- Wei Yan
- Laboratory of Natural and targeted small molecule drugs, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yong Li
- Laboratory of Natural and targeted small molecule drugs, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yan Liu
- Laboratory of Natural and targeted small molecule drugs, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yi Wen
- Laboratory of Natural and targeted small molecule drugs, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Heying Pei
- Laboratory of Natural and targeted small molecule drugs, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Jianhong Yang
- Laboratory of Natural and targeted small molecule drugs, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China.
| | - Lijuan Chen
- Laboratory of Natural and targeted small molecule drugs, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China.
| |
Collapse
|
169
|
Anker AM, Lenhard J, Klein SM, Felthaus O, Prantl L, Ruewe M. Standard doses of Triamcinolone do not affect fibroblast cell migration of abdominoplasty patients in-vitro1. Clin Hemorheol Microcirc 2023; 85:307-314. [PMID: 36502314 DOI: 10.3233/ch-229106] [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] [Indexed: 12/12/2022]
Abstract
BACKGROUND Recent studies have demonstrated that local application of corticosteroids reduces wound exudation following abdominoplasty and other reconstructive surgical procedures. On the other hand, corticosteroids might provoke wound healing disturbances due to their immunosuppressive effects. OBJECTIVE The main objective of this study was to gain further information about the impact of the corticosteroid triamcinolone on cell migration in abdominoplasty patients. METHODS An in-vitro scratch assay wound healing model was applied to observe cell migration of fibroblasts cultured with nutrient medium containing human seroma aspirate±triamcinolone. RESULTS There were no significant differences regarding cell migration when fibroblasts were incubated with triamcinolone + seroma containing culture medium compared to seroma containing culture medium without triamcinolone. CONCLUSIONS The performed in-vitro study suggests that triamcinolone does not decelerate fibroblast cell migration which is considered as a surrogate of wound healing.
Collapse
Affiliation(s)
- Alexandra M Anker
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Regensburg and Caritas Hospital St. Josef Regensburg, Regensburg, Germany
| | - Jasmin Lenhard
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Regensburg and Caritas Hospital St. Josef Regensburg, Regensburg, Germany
| | - Silvan M Klein
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Regensburg and Caritas Hospital St. Josef Regensburg, Regensburg, Germany
| | - Oliver Felthaus
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Regensburg and Caritas Hospital St. Josef Regensburg, Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Regensburg and Caritas Hospital St. Josef Regensburg, Regensburg, Germany
| | - Marc Ruewe
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Regensburg and Caritas Hospital St. Josef Regensburg, Regensburg, Germany
| |
Collapse
|
170
|
Protective role of Decylubiquinone against secondary melanoma at lung in B16F10 induced mice by reducing E-cadherin expression and ameliorating ROCKII-Limk1/2-Cofiliin mediated metastasis. Cell Signal 2023; 101:110486. [PMID: 36208704 DOI: 10.1016/j.cellsig.2022.110486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
Melanoma is one of the most consequential skin cancer with a rising death incidences. Silent but belligerent nature of metastatic sprouting is the leading cause of melanoma related mortality. Invasion of metastatic cells and re-expression of E-Cadherin play the crucial role in the establishment of secondary tumor at distal sites. Thus, manipulation of tumor cell invasion in parallel to regulation of E-Cadherin expression can be considered as potential anti-metastatic strategy. Evidences suggested key role of reactive oxygen species associated ROCK activities in the modulation of metastatic invasion via F-actin stabilization. Here, we first-time report Decylubiquinone, a dietary Coenzyme Q10 analog, as an effective attenuator of pulmonary metastatic melanoma in C57BL/6 mice. Current study depicted detailed molecular interplay associated with Decylubiquinone mediated phosphorylation of ROCKII at Tyr722 along with reduced phosphorylation of ROCKII Ser1366 leading to suppression of Limk1/2-Cofilin-F-actin stabilization axis that finally restricted B16F10 melanoma cell invasion at metastatic site. Analysis further deciphered the role of HNF4α as its nuclear translocation modulated E-Cadherin expression, the effect of reactive oxygen species dependent ROCKII activity in secondarily colonized B16F10 melanoma cells at lungs. Thus unbosoming of related signal orchestra represented Decylubiquinone as a potential remedial agent against secondary lung melanoma.
Collapse
|
171
|
Rhoda C, Sunda F, Kidzeru E, Khumalo NP, Arowolo A. FAM111B dysregulation promotes malignancy in fibrosarcoma and POIKTMP and a low-cost method for its mutation screening. Cancer Treat Res Commun 2023; 34:100679. [PMID: 36610347 DOI: 10.1016/j.ctarc.2022.100679] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/05/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Mutations in the uncharacterised human FAM111B gene are associated with POIKTMP, a rare multi-organ fibrosing disease. Recent studies also reported the overexpression of FAM111B in specific cancers. Moreover, FAM111B mutation screening may prove expensive in under-resourced facilities. Therefore, this study investigated its cellular function and dysfunction and described an inexpensive mutation screening method. MATERIALS AND METHODS FAM111B expression was assessed in silico and validated in vitro in cell lines and primary skin fibroblasts from a South African POIKTMP-patient with the heterozygous FAM111B gene mutation: NM_198947.4: c.1861T>G (p. Tyr621Asp or Y621D) by qPCR and western blot. The cellular function of FAM111B was studied in HT1080 using various cell-based functional assays, and the Y621D mutation was genotyped by PCR-RFLP. RESULTS Expression studies showed upregulated FAM111B mRNA and protein in the cancer cells. High FAM111B expression with robust nuclear localization occurred in HT1080. Additionally, expression data and cell-based assays indicated that FAM111B led to the upregulation of cell migration, decreased cell apoptosis, and modulatory effects on cell proliferation. Y621D mutation showed similar effects on cell migration but minimal impact on cell apoptosis. FAM111B mRNA and protein expression were markedly downregulated (p ≤ 0.05) in the POIKTMP-patient's fibroblasts. The PCR-RFLP method successfully genotyped Y621D gene mutation. DISCUSSION FAM111B is a cancer-associated nuclear protein: Its modulation by mutations or overexpression may contribute to the malignancy of cancers and POIKTMP/fibrosis and poor clinical outcomes and represents a viable prognostic marker or therapeutic target. Furthermore, the PCR-RFLP method could prove a valuable tool for FAM111B mutation validation or screening in resource-constrained laboratories.
Collapse
Affiliation(s)
- Cenza Rhoda
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Falone Sunda
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Elvis Kidzeru
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Nonhlanhla P Khumalo
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Afolake Arowolo
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa.
| |
Collapse
|
172
|
Eissa IH, Elkaeed EB, Elkady H, Yousef RG, Alsfouk BA, Elzahabi HSA, Ibrahim IM, Metwaly AM, Husein DZ. Design, Molecular Modeling, MD Simulations, Essential Dynamics, ADMET, DFT, Synthesis, Anti-proliferative, and Apoptotic Evaluations of a New Anti-VEGFR-2 Nicotinamide Analogue. Curr Pharm Des 2023; 29:2902-2920. [PMID: 38031271 DOI: 10.2174/0113816128274870231102114858] [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: 08/20/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023]
Abstract
OBJECTIVES This study aims to design and evaluate (in silico and in vitro) a new nicotinamide derivative as an inhibitor of VEGFR-2, a major mediator of angiogenesis Methods: The following in silico studies were performed; DFT calculations, molecular modelling, MD simulations, MM-GBSA, PLIP, and PCAT studies. The compound's in silico (ADMET) analysis was also conducted. Subsequently, the compound ((E)-N-(4-(1-(2-(4-(4-Chlorobenzamido)benzoyl)hydrazono)ethyl) phenyl)nicotinamide) was successfully synthesized and designated as compound X. In vitro, VEGFR-2 inhibition and cytotoxicity of compound X against HCT-116 and A549 cancer cell lines and normal Vero cell lines were conducted. Apoptosis induction and migration assay of HCT-116 cell lines after treatment with compound X were also evaluated. RESULTS DFT calculations assigned stability and reactivity of compound X. Molecular docking and MD simulations indicated its excellent binding against VEGFR-2. Furthermore, MM-GBSA analysis, PLIP experiments, and PCAT studies confirmed compound X's correct binding with optimal dynamics and energy. ADMET analysis expressed its general likeness and safety. The in vitro assays demonstrated that compound X effectively inhibited VEGFR-2, with an IC50 value of 0.319 ± 0.013 μM and displayed cytotoxicity against HCT-116 and A549 cancer cell lines, with IC50 values of 57.93 and 78.82 μM, respectively. Importantly, compound X exhibited minimal toxicity towards the non-cancerous Vero cell lines, (IC50 = 164.12 μM). Additionally, compound X significantly induced apoptosis of HCT-116 cell lines and inhibited their potential to migrate and heal. CONCLUSION In summary, the presented study has identified compound X as a promising candidate for the development of a novel apoptotic lead anticancer drug.
Collapse
Affiliation(s)
- Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Eslam B Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh 13713, Saudi Arabia
| | - Hazem Elkady
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Reda G Yousef
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Bshra A Alsfouk
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Heba S A Elzahabi
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11884, Egypt
| | - Ibrahim M Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Ahmed M Metwaly
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
- Biopharmaceutical Products Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Alexandria, Egypt
| | - Dalal Z Husein
- Chemistry Department, Faculty of Science, New Valley University, El-Kharja 72511, Egypt
| |
Collapse
|
173
|
Revati R, Majid MSA, Ridzuan MJM, Mamat N, Cheng EM, Alshahrani HA. In vitro biodegradation, cytotoxicity, and biocompatibility of polylactic acid/napier cellulose nanofiber scaffold composites. Int J Biol Macromol 2022; 223:479-489. [PMID: 36368357 DOI: 10.1016/j.ijbiomac.2022.11.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/04/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022]
Abstract
This study aimed to evaluate the bioactivities and biocompatibilities of porous polylactic acid (PLA) reinforced with cellulose nanofiber (CNF) scaffolds. The in vitro degradation behaviors of the porous PLA/CNF scaffolds were systematically measured for up to 8 weeks in a phosphate-buffered saline medium at 37 °C. The reinforcement of CNF resisted the biodegradation of the scaffolds. The in vitro cytotoxicity and biocompatibility of the scaffolds were determined using the Beas2B American Type Culture Collection cells. The 3-(4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide cytotoxicity and proliferation tests showed that the scaffolds were non-toxic, and epithelial cells grew well on the scaffold after 7 days of culture, whereas the percentage of cell proliferation on the PLA/CNF15 scaffold was the largest, 130 %. A scratch wound-healing assay was performed to evaluate the suitability of the scaffolds for cell migration. The results demonstrated that the scaffolds exhibited good cell migration towards nearly complete wound closure.
Collapse
Affiliation(s)
- R Revati
- Faculty of Mechanical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Putra Campus, Arau 02600, Perlis, Malaysia; Faculty of Electronic Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Putra Campus, Arau 02600, Perlis, Malaysia
| | - M S Abdul Majid
- Faculty of Mechanical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Putra Campus, Arau 02600, Perlis, Malaysia.
| | - M J M Ridzuan
- Faculty of Mechanical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Putra Campus, Arau 02600, Perlis, Malaysia
| | - N Mamat
- Faculty of Electronic Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Putra Campus, Arau 02600, Perlis, Malaysia
| | - E M Cheng
- Faculty of Electronic Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Putra Campus, Arau 02600, Perlis, Malaysia
| | - Hassan A Alshahrani
- Department of Mechanical Engineering, College of Engineering, Najran University, Najran 11001, Saudi Arabia
| |
Collapse
|
174
|
Aly SH, El-Hassab MA, Elhady SS, Gad HA. Comparative Metabolic Study of Tamarindus indica L.'s Various Organs Based on GC/MS Analysis, In Silico and In Vitro Anti-Inflammatory and Wound Healing Activities. PLANTS (BASEL, SWITZERLAND) 2022; 12:87. [PMID: 36616217 PMCID: PMC9824397 DOI: 10.3390/plants12010087] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 06/17/2023]
Abstract
The chemical composition of the n-hexane extract of Tamarindus indica's various organs-bark, leaves, seeds, and fruits (TIB, TIL, TIS, TIF)-was investigated using gas chromatography-mass spectrometry (GC/MS) analysis. A total of 113 metabolites were identified, accounting for 93.07, 83.17, 84.05, and 85.08 % of the total identified components in TIB, TIL, TIS, and TIF, respectively. Lupeol was the most predominant component in TIB and TIL, accounting for 23.61 and 22.78%, respectively. However, n-Docosanoic acid (10.49%) and methyl tricosanoate (7.09%) were present in a high percentage in TIS. However, α-terpinyl acetate (7.36%) and α-muurolene (7.52%) were the major components of TIF n-hexane extract. By applying a principal component analysis (PCA) and hierarchal cluster analysis (HCA) to GC/MS-based metabolites, a clear differentiation of Tamarindus indica organs was achieved. The anti-inflammatory activity was evaluated in vitro on lipopolysaccharide (LPS)-induced RAW 264.7 macrophages. In addition, the wound healing potential for the n-hexane extract of various plant organs was assessed using the in-vitro wound scratch assay using Human Skin Fibroblast cells. The tested extracts showed considerable anti-inflammatory and wound-healing activities. At a concentration of 10 µg/mL, TIL showed the highest nitric oxide (NO) inhibition by 53.97 ± 5.89%. Regarding the wound healing potential, after 24 h, TIB, TIL, TIS, and TIF n-hexane extracts at 10 g/mL reduced the wound width to 1.09 ± 0.04, 1.12 ± 0.18, 1.09 ± 0.28, and 1.41 ± 0.35 mm, respectively, as compared to the control cells (1.37 ± 0.15 mm). These findings showed that the n-hexane extract of T. indica enhanced wound healing by promoting fibroblast migration. Additionally, a docking study was conducted to assess the major identified phytoconstituents' affinity for binding to glycogen synthase kinase 3-β (GSK3-β), matrix metalloproteinases-8 (MMP-8), and nitric oxide synthase (iNOS). Lupeol showed the most favourable binding affinity to GSK3-β and iNOS, equal to -12.5 and -13.7 Kcal/mol, respectively, while methyl tricosanoate showed the highest binding affinity with MMP-8 equal to -13.1 Kcal/mol. Accordingly, the n-hexane extract of T. indica's various organs can be considered a good candidate for the management of wound healing and inflammatory conditions.
Collapse
Affiliation(s)
- Shaza H. Aly
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo, Cairo 11829, Egypt
| | - Mahmoud A. El-Hassab
- Department of Medicinal Chemistry, Faculty of Pharmacy, King Salman International University (KSIU), South Sinai 46612, Egypt
| | - Sameh S. Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Haidy A. Gad
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, King Salman International University (KSIU), South Sinai 46612, Egypt
| |
Collapse
|
175
|
Ahmad N. In Vitro and In Vivo Characterization Methods for Evaluation of Modern Wound Dressings. Pharmaceutics 2022; 15:42. [PMID: 36678671 PMCID: PMC9864730 DOI: 10.3390/pharmaceutics15010042] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/10/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022] Open
Abstract
Chronic wound management represents a major challenge in the healthcare sector owing to its delayed wound-healing process progression and huge financial burden. In this regard, wound dressings provide an appropriate platform for facilitating wound healing for several decades. However, adherent traditional wound dressings do not provide effective wound healing for highly exudating chronic wounds and need the development of newer and innovative wound dressings to facilitate accelerated wound healing. In addition, these dressings need frequent changing, resulting in more pain and discomfort. In order to overcome these issues, a wide range of affordable and innovative modern wound dressings have been developed and explored recently to accelerate and improve the wound healing process. However, a comprehensive understanding of various in vitro and in vivo characterization methods being utilized for the evaluation of different modern wound dressings is lacking. In this context, an overview of modern dressings and their complete in vitro and in vivo characterization methods for wound healing assessment is provided in this review. Herein, various emerging modern wound dressings with advantages and challenges have also been reviewed. Furthermore, different in vitro wound healing assays and in vivo wound models being utilized for the evaluation of wound healing progression and wound healing rate using wound dressings are discussed in detail. Finally, a summary of modern wound dressings with challenges and the future outlook is highlighted.
Collapse
Affiliation(s)
- Naveed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| |
Collapse
|
176
|
Bonaterra GA, Schmitt J, Schneider K, Schwarzbach H, Aziz-Kalbhenn H, Kelber O, Müller J, Kinscherf R. Phytohustil ® and root extract of Althaea officinalis L. exert anti-inflammatory and anti-oxidative properties and improve the migratory capacity of endothelial cells in vitro. Front Pharmacol 2022; 13:948248. [PMID: 36569306 PMCID: PMC9773075 DOI: 10.3389/fphar.2022.948248] [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: 05/19/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction: Althaea officinalis L.'s root extract (REA) has been used as a medicinal plant since ancient times to treat a cough. Applying REA leads to a protective film that induces a faster regeneration of the lesioned laryngopharyngeal mucosa caused by dry coughs. The buccopharyngeal mucosa is a highly vascularized tissue. In this regard, anti-inflammatory/-oxidant phytochemicals that improve the repair of the lesion site, e.g., neovascularization in the wound, are critical for promoting healing. For this reason, it is essential to investigate the effects of Phytohustil® and REA on different cellular components of the mucosa under conditions similar to those found in the injured mucosa. Thus, this in vitro study investigated the anti-inflammatory/oxidative and pro-migratory properties of Phytohustil® cough syrup on vascular endothelial cells. Methods: Human umbilical vein endothelial cells (HUVEC) were pretreated (24 h) with Phytohustil®, its excipients, or REA, followed by incubation with hydrogen peroxide (H2O2; 1 h; pro-oxidative) or with lipopolysaccharides (LPS; 3 h; pro-inflammatory). Viability and cytotoxicity were measured by PrestoBlue® assay. Intracellular reactive oxygen species (ROS) were quantified with 20-70-dichlorofluorescein diacetate (DCFDA). The release of interleukin 6 (IL6) was determined by enzyme-linked immunosorbent assay (ELISA). The migratory capacity of HUVEC was measured using a scratch assay. Results: Our results show that Phytohustil®, its excipients and REA were not cytotoxic. Pretreatment of HUVEC (24 h) with Phytohustil® or REA inhibited the LPS-activated IL6 release. Phytohustil® or REA inhibited the H2O2-induced cytotoxicity and intracellular ROS production. Phytohustil® and REA significantly stimulated wound closure compared to the control. Conclusion: Our data show that Phytohustil® and REA have anti-inflammatory/-oxidant properties and improve the migratory capacity of vascular endothelial cells. These properties may contribute to the healing characteristics of Phytohustil® and support the benefit of Phytohustil® in patient's treatment of irritated oral mucosa.
Collapse
Affiliation(s)
- Gabriel A. Bonaterra
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany,*Correspondence: Gabriel A. Bonaterra,
| | - Johanna Schmitt
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Kim Schneider
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Hans Schwarzbach
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Heba Aziz-Kalbhenn
- Bayer Consumer Health Division, Phytomedicines Supply and Development Center, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Olaf Kelber
- Bayer Consumer Health Division, Phytomedicines Supply and Development Center, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Jürgen Müller
- Bayer Consumer Health Division, Phytomedicines Supply and Development Center, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Ralf Kinscherf
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| |
Collapse
|
177
|
Ma H, Ge P, Bian S, Li Y, Ni A, Zhang R, Wang Y, Zhao J, Zong Y, Yuan J, Sun Y, Chen J. miR-193-5p negatively regulates PIK3CD to promote crop fibrocyte proliferation in pigeon (Columba livia). Poult Sci 2022; 102:102378. [PMID: 36565634 PMCID: PMC9801189 DOI: 10.1016/j.psj.2022.102378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/13/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
The crop of pigeon has specific characteristics as producing crop milk in the lactating period. However, the exact mechanisms underlying the regulation of crop lactation remain unclear. miRNAs, the essential regulators of gene expression, are implicated in various physiological and biological activities. In this study, we discovered a new miRNA that regulated phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta (PIK3CD) and crop fibrocyte proliferation. Results of the luciferase reporter assay suggested that miR-193-5p suppressed PIK3CD expression by targeting a conserved binding site in the 3'-untranslated region (UTR) of PIK3CD mRNA. MiR-193-5p promoted crop fibrocyte proliferation and migration, whereas PIK3CD inhibited these effects. These findings suggested an important regulatory role of miR-193-5p in crop fibrocyte proliferation, suggesting that miR-193-5p and PIK3CD might be important regulators of crop milk production.
Collapse
|
178
|
Nitta RT, Luo EJ, Lim M, Li G. Can tumor treating fields induce DNA damage and reduce cell motility in medulloblastoma cell lines? J Neurosurg Pediatr 2022; 30:555-566. [PMID: 36208441 DOI: 10.3171/2022.8.peds22300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/25/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Medulloblastoma (MB) is the most common malignant pediatric brain tumor and accounts for approximately 20% of all pediatric CNS tumors. Current multimodal treatment is associated with a 70%-90% 5-year survival rate; however, the prognosis for patients with tumor dissemination and recurrent MB remains poor. The majority of survivors exhibit long-term neurocognitive complications; thus, more effective and less toxic treatments are critically needed. Tumor treating fields (TTFields) are low-intensity, alternating electric fields that disrupt cell division through physical interactions with key molecules during mitosis. Side effects from TTField therapy are minimal, making it an ideal candidate for MB treatment. METHODS To determine if TTFields can be an effective treatment for MB, the authors conducted an in vitro study treating multiple MB cell lines. Three MB molecular subgroups (SHH [sonic hedgehog], group 3, and group 4) were treated for 24, 48, and 72 hours at 100, 200, 300, and 400 kHz. Combinatorial studies were conducted with the small-molecule casein kinase 2 inhibitor CX-4945. RESULTS TTFields reduced MB cell growth with an optimal frequency of 300 kHz, and the most efficacious treatment time was 72 hours. Treatment with TTFields dysregulated actin polymerization and corresponded with a reduction in cell motility and invasion. TTFields also induced DNA damage (γH2AX, 53BP1) that correlated with an increase in apoptotic cells. The authors discovered that CX-4945 works synergistically with TTFields to reduce MB growth. In addition, combining CX-4945 and TTFields increased the cellular actin dysregulation, which correlated with a decrease in MB migration. CONCLUSIONS The findings of this study demonstrate that TTFields may be a novel and less toxic method to treat patients with MB.
Collapse
|
179
|
Jothimani G, Ganesan H, Pathak S, Banerjee A. Molecular Characterization of Primary and Metastatic Colon Cancer Cells to Identify Therapeutic Targets with Natural Compounds. Curr Top Med Chem 2022; 22:2598-2615. [PMID: 35366775 DOI: 10.2174/1568026622666220401161511] [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: 09/30/2021] [Revised: 02/01/2022] [Accepted: 02/10/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND Metastasis is the world's leading cause of colon cancer morbidity. Due to its heterogeneity, it has been challenging to understand primary to metastatic colon cancer progression and find a molecular target for colon cancer treatment. OBJECTIVES The current investigation aimed to characterize the immune and genotypic profiles of primary and metastatic colon cancer cell lines and identify a molecular target for colon cancer treatment. METHODS Colony-forming potential, migration and invasion potential, cytokine profiling, miRNA, and mRNA expression were examined. Molecular docking for the Wnt signaling proteins with various plant compounds was performed. RESULTS Colony formation, migration, and invasion potential were significantly higher in metastatic cells. The primary and metastatic cells' local immune and genetic status revealed TGF β-1, IL-8, MIP-1b, I-TAC, GM-CSF, and MCP-1 were highly expressed in all cancer cells. RANTES, IL-4, IL- 6, IFNγ, and G-CSF were less expressed in cancer cell lines. mRNA expression analysis displayed significant overexpression of proliferation, cell cycle, and oncogenes, whereas apoptosis cascade and tumor suppressor genes were significantly down-regulated in metastatic cells more evidently. Most importantly, the results of molecular docking with dysregulated Wnt signaling proteins shows that peptide AGAP and coronaridine had maximum hydrogen bonds to β-catenin and GSK3β with a better binding affinity. CONCLUSION This study emphasized genotypic differences between the primary and metastatic colon cancer cells, delineating the intricate mechanisms to understand the primary to metastatic advancement. The molecular docking aided in understanding the future molecular targets for bioactive- based colon cancer therapeutic interventions.
Collapse
Affiliation(s)
- Ganesan Jothimani
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| | - Harsha Ganesan
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| |
Collapse
|
180
|
Küllmer F, Vepřek NA, Borowiak M, Nasufović V, Barutzki S, Thorn-Seshold O, Arndt HD, Trauner D. Next Generation Opto-Jasplakinolides Enable Local Remodeling of Actin Networks. Angew Chem Int Ed Engl 2022; 61:e202210220. [PMID: 36048143 PMCID: PMC11256906 DOI: 10.1002/anie.202210220] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Indexed: 11/12/2022]
Abstract
The natural product jasplakinolide is widely used to stabilize F-actin. Based on extensive structure-activity relationship studies, we have developed a new generation of photoswitchable jasplakinolides that feature rationally designed red-shifted azobenzene photoswitches. Our lead compound, nOJ, can be activated with longer wavelengths in the visible range (e.g. 440-475 nm) and rapidly returns to its inactive state through thermal relaxation. nOJ enables the reversible control of F-actin dynamics, as shown through live-cell imaging, cell migration, and cell proliferation assays. Short, local irradiation with blue light resulted in highly localized and reversible actin aggregation with subcellular precision. Our optical tool can be useful in diverse fields to study actin dynamics with excellent spatiotemporal resolution.
Collapse
Affiliation(s)
- Florian Küllmer
- Friedrich-Schiller-Universität (FSU), Institut für Organische Chemie und Makromolekulare Chemie, Humboldtstr. 10, 07743, Jena, Germany
| | - Nynke A Vepřek
- New York University, Department of Chemistry, 100 Washington Square East, New York, NY 10003, USA
- Department of Chemistry, Ludwig Maximilian University of Munich, Butenandtstrasse 5-13, 81377, München, Germany
| | - Malgorzata Borowiak
- Department of Pharmacy, Ludwig Maximilian University of Munich, Butenandtstrasse 5-13, 81377, München, Germany
| | - Veselin Nasufović
- Friedrich-Schiller-Universität (FSU), Institut für Organische Chemie und Makromolekulare Chemie, Humboldtstr. 10, 07743, Jena, Germany
| | - Sebastian Barutzki
- Friedrich-Schiller-Universität (FSU), Institut für Organische Chemie und Makromolekulare Chemie, Humboldtstr. 10, 07743, Jena, Germany
| | - Oliver Thorn-Seshold
- Department of Pharmacy, Ludwig Maximilian University of Munich, Butenandtstrasse 5-13, 81377, München, Germany
| | - Hans-Dieter Arndt
- Friedrich-Schiller-Universität (FSU), Institut für Organische Chemie und Makromolekulare Chemie, Humboldtstr. 10, 07743, Jena, Germany
| | - Dirk Trauner
- New York University, Department of Chemistry, 100 Washington Square East, New York, NY 10003, USA
- Department of Chemistry University of Pennsylvania, 231 South 34th St., Philadelphia, PA 19104-6323, USA
| |
Collapse
|
181
|
Gaertner K, Michell C, Tapanainen R, Goffart S, Saari S, Soininmäki M, Dufour E, Pohjoismäki JLO. Molecular phenotyping uncovers differences in basic housekeeping functions among closely related species of hares (
Lepus
spp., Lagomorpha: Leporidae). Mol Ecol 2022. [PMID: 36320183 DOI: 10.1111/mec.16755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/16/2022] [Accepted: 10/06/2022] [Indexed: 11/15/2022]
Abstract
Speciation is a fundamental evolutionary process, which results in genetic differentiation of populations and manifests as discrete morphological, physiological and behavioural differences. Each species has travelled its own evolutionary trajectory, influenced by random drift and driven by various types of natural selection, making the association of genetic differences between the species with the phenotypic differences extremely complex to dissect. In the present study, we have used an in vitro model to analyse in depth the genetic and gene regulation differences between fibroblasts of two closely related mammals, the arctic/subarctic mountain hare (Lepus timidus Linnaeus) and the temperate steppe-climate adapted brown hare (Lepus europaeus Pallas). We discovered the existence of a species-specific expression pattern of 1623 genes, manifesting in differences in cell growth, cell cycle control, respiration, and metabolism. Interspecific differences in the housekeeping functions of fibroblast cells suggest that speciation acts on fundamental cellular processes, even in these two interfertile species. Our results help to understand the molecular constituents of a species difference on a cellular level, which could contribute to the maintenance of the species boundary.
Collapse
Affiliation(s)
- Kateryna Gaertner
- Mitochondrial Bioenergetics and Metabolism, Faculty of Medicine and Health Technology FI‐33014 Tampere University Tampere Finland
| | - Craig Michell
- Department of Environmental and Biological Sciences FI‐80101 University of Eastern Finland Kuopio Finland
- Red Sea Research Center, Division of Biological and Environmental Science and Engineering King Abdullah University of Science and Technology (KAUST) Thuwal Saudi Arabia
| | - Riikka Tapanainen
- Department of Environmental and Biological Sciences FI‐80101 University of Eastern Finland Kuopio Finland
| | - Steffi Goffart
- Department of Environmental and Biological Sciences FI‐80101 University of Eastern Finland Kuopio Finland
| | - Sina Saari
- Mitochondrial Bioenergetics and Metabolism, Faculty of Medicine and Health Technology FI‐33014 Tampere University Tampere Finland
| | - Manu Soininmäki
- Department of Environmental and Biological Sciences FI‐80101 University of Eastern Finland Kuopio Finland
| | - Eric Dufour
- Mitochondrial Bioenergetics and Metabolism, Faculty of Medicine and Health Technology FI‐33014 Tampere University Tampere Finland
| | - Jaakko L. O. Pohjoismäki
- Department of Environmental and Biological Sciences FI‐80101 University of Eastern Finland Kuopio Finland
| |
Collapse
|
182
|
Aiyer A, Das T, Whiteley GS, Glasbey T, Kriel FH, Farrell J, Manos J. The Efficacy of an N-Acetylcysteine-Antibiotic Combination Therapy on Achromobacter xylosoxidans in a Cystic Fibrosis Sputum/Lung Cell Model. Biomedicines 2022; 10:2886. [PMID: 36359406 PMCID: PMC9687303 DOI: 10.3390/biomedicines10112886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 09/29/2023] Open
Abstract
Cystic fibrosis (CF) is a disorder causing dysfunctional ion transport resulting in the accumulation of viscous mucus. This environment fosters a chronic bacterial biofilm-associated infection in the airways. Achromobacter xylosoxidans, a gram-negative aerobic bacillus, has been increasingly associated with antibiotic resistance and chronic colonisation in CF. In this study, we aimed to create a reproducible model of CF infection using an artificial sputum medium (ASMDM-1) with bronchial (BEAS-2B) and macrophage (THP-1) cells to test A. xylosoxidans infection and treatment toxicity. This study was conducted in three distinct stages. First, the tolerance of BEAS-2B cell lines and two A. xylosoxidans strains against ASMDM-1 was optimised. Secondly, the cytotoxicity of combined therapy (CT) comprising N-acetylcysteine (NAC) and the antibiotics colistin or ciprofloxacin was tested on cells alone in the sputum model in both BEAS-2B and THP-1 cells. Third, the efficacy of CT was assessed in the context of a bacterial infection within the live cell/sputum model. We found that a model using 20% ASMDM-1 in both cell populations tolerated a colistin-NAC-based CT and could significantly reduce bacterial loads in vitro (~2 log10 CFU/mL compared to untreated controls). This pilot study provides the foundation to study other bacterial opportunists that infect the CF lung to observe infection and CT kinetics. This model also acts as a springboard for more complex co-culture models.
Collapse
Affiliation(s)
- Aditi Aiyer
- Charles Perkins Centre, Infection, Immunity and Inflammation, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Theerthankar Das
- Charles Perkins Centre, Infection, Immunity and Inflammation, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Gregory S. Whiteley
- Charles Perkins Centre, Infection, Immunity and Inflammation, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Whiteley Corporation, Level 5, 12 Mount Street North Sydney, Sydney, NSW 2060, Australia
- School of Medicine, Western Sydney University, Sydney, NSW 2566, Australia
| | - Trevor Glasbey
- Whiteley Corporation, 19-23 Laverick Avenue, Tomago, NSW 2322, Australia
| | - Frederik H. Kriel
- Whiteley Corporation, 19-23 Laverick Avenue, Tomago, NSW 2322, Australia
| | - Jessica Farrell
- Charles Perkins Centre, Infection, Immunity and Inflammation, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Whiteley Corporation, Level 5, 12 Mount Street North Sydney, Sydney, NSW 2060, Australia
| | - Jim Manos
- Charles Perkins Centre, Infection, Immunity and Inflammation, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
183
|
Li W, Shi C, Wu X, Zhang Y, Liu H, Wang X, Huang C, Liang L, Liu Y. Light activation of iridium(III) complexes driving ROS production and DNA damage enhances anticancer activity in A549 cells. J Inorg Biochem 2022; 236:111977. [PMID: 36030672 DOI: 10.1016/j.jinorgbio.2022.111977] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/10/2022] [Accepted: 08/19/2022] [Indexed: 12/15/2022]
Abstract
The work aimed to synthesize and characterize two iridium(III) complexes [Ir(ppy)2(IPPH)](PF6) (Ir1, IPPH = (2S,3R,5S,6R)-2-(2-(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)phenoxy)-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol, ppy = 2-phenylpyridine), [Ir(piq)2(IPPH)](PF6) (Ir2, piq = 1-phenylisoquinoline). The cytotoxicity of the complexes against BEL-7402, A549, HCT-116, B16 cancer cells and normal LO2 was evaluated through 3-(4,5-dimethylthiazole-2-yl)-2,5-biphenyl tetrazolium bromide (MTT) method. The complexes show no cytotoxic activity (IC50 > 100 μM) against these cancer cells, while their cytotoxicity can significantly be elevated upon illumination. The IC50 values range from 0.2 ± 0.05 to 35.5 ± 3.5 μM. The cellular uptake, endoplasmic reticulum and mitochondria localization, reactive oxygen species, the change of mitochondrial membrane potential, γ-H2AX levels, cycle arrest, apoptosis and the expression of B-cell lymphoma-2 were investigated. The calreticulin (CRT), heat shock protein 70 (HSP70), high mobility group box 1 (HMGB1) were explored. This study demonstrates that photoactivatable complexes induce cell death in A549 through ROS-mediated endoplasmic reticulum stress-mitochondrial pathway, DNA damage pathways, immunogenic cell death (ICD), activation of PI3K/AKT signaling pathway and inhibit the cell growth at S phase.
Collapse
Affiliation(s)
- Wenlong Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Chuanling Shi
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Xiaoyun Wu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yuanyuan Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Haimei Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Xiuzhen Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| | - Chunxia Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lijuan Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems and Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| |
Collapse
|
184
|
Speciation Analysis Highlights the Interactions of Auranofin with the Cytoskeleton Proteins of Lung Cancer Cells. Pharmaceuticals (Basel) 2022; 15:ph15101285. [PMID: 36297397 PMCID: PMC9610265 DOI: 10.3390/ph15101285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 12/01/2022] Open
Abstract
Two types of lung cells (epithelial cancer lung cells, A-549 and lung fibroblasts MRC-5) were exposed to the clinically established gold drug auranofin at concentrations close to the half-maximal inhibitory drug concentrations (IC50). Collected cells were subjected to speciation analysis using inductively coupled plasma mass spectrometry (ICP-MS). Auranofin showed better affinity toward proteins than DNA, RNA, and hydrophilic small molecular weight compounds. It can bind to proteins that vary in size (~20 kDa, ~75 kDa, and ≥200 kDa) and pI. However, the possibility of dimerization and protein–protein complex formation should also be taken into account. µRPLC/CZE-ESI-MS/MS studies on trypsinized proteins allowed the indication of 76 peptides for which signal intensity was influenced by auranofin presence in cells. Based on it, identity was proposed for 20 proteins. Except for thioredoxin reductase (TrxR), which is directly targeted by gold complex, the proteins were found to be transformed. Five indicated proteins: myosin, plectin, talin, two annexins, and kinase M3K5, are responsible for cell–cell, cell–protein interactions, and cell motility. A wound healing test confirmed their regulation by auranofin as cell migration decreased by 40% while the cell cycle was not interrupted.
Collapse
|
185
|
Piet M, Paduch R. Ursolic and oleanolic acids in combination therapy inhibit migration of colon cancer cells through down-regulation of the uPA/uPAR-dependent MMPs pathway. Chem Biol Interact 2022; 368:110202. [PMID: 36191607 DOI: 10.1016/j.cbi.2022.110202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/12/2022] [Accepted: 09/21/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Colorectal cancer is one the most lethal cancers worldwide. Since chemotherapy is burdened with harmful effects, agents capable of enhancing the chemotherapeutic effect are being sought. Ursolic acid (UA) and oleanolic acid (OA) were analyzed for such properties. The aim of the study was to evaluate the ability of UA and OA administered individually and in combination with each other and/or a cytostatic drug camptothecin-11 (CPT-11) to limit the viability and migration of colorectal cancer cells. MATERIALS AND METHODS The cytotoxic effect of UA, OA and CPT-11 and impact on normal and cancer cell migration rate were assessed. Furthermore, the effect on factors crucial in cancer metastasis: MMP-2 and -9, uPA/uPAR, and E-cadherin were assessed with ELISA, Western Blotting and immunofluorescence assays. Statistical analysis was performed with One-Way Anova with Dunnett's test. RESULTS The studied compounds exhibited the most favorable properties, i.e. they reduced the viability and migration of cancer cells. Furthermore, the secretion, activity, and cellular level of cancer MMP-2 and -9 were decreased, as a result of uPA/uPAR down-regulation. The agents also increased the level of cellular E-cadherin. The effect of the studied agents on normal cells was milder. CONCLUSIONS The compounds exhibited stronger activity when administered in combination and, combined with CPT-11, enhanced anti-tumorigenic activity of the drug. The migration-limiting activity was based on down-regulation of the uPA/uPAR-dependent MMP pathway. Moreover, UA and OA exhibited a protective effect towards normal cells.
Collapse
Affiliation(s)
- Mateusz Piet
- Department of Virology and Immunology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Roman Paduch
- Department of Virology and Immunology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland; Department of General Ophthalmology, Faculty of Medicine, Medical University of Lublin, ul Chmielna 1, 20-079, Lublin, Poland.
| |
Collapse
|
186
|
Nawaz A, Zaman Safi S, Sikandar S, Zeeshan R, Zulfiqar S, Mehmood N, Alobaid HM, Rehman F, Imran M, Tariq M, Ali A, Emran TB, Yar M. Heparin-Loaded Alginate Hydrogels: Characterization and Molecular Mechanisms of Their Angiogenic and Anti-Microbial Potential. MATERIALS (BASEL, SWITZERLAND) 2022; 15:6683. [PMID: 36234025 PMCID: PMC9573464 DOI: 10.3390/ma15196683] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 05/06/2023]
Abstract
Background: Chronic wounds continue to be a global concern that demands substantial resources from the healthcare system. The process of cutaneous wound healing is complex, involving inflammation, blood clotting, angiogenesis, migration and remodeling. In the present study, commercially available alginate wound dressings were loaded with heparin. The purpose of the study was to enhance the angiogenic potential of alginate wound dressings and analyze the antibacterial activity, biocompatibility and other relevant properties. We also aimed to conduct some molecular and gene expression studies to elaborate on the mechanisms through which heparin induces angiogenesis. Methods: The physical properties of the hydrogels were evaluated by Fourier transform infrared spectroscopy (FTIR). Swelling ability was measured by soaking hydrogels in the Phosphate buffer at 37 °C, and cell studies were conducted to evaluate the cytotoxicity and biocompatibility of hydrogels in NIH3T3 (fibroblasts). Real-time PCR was conducted to check the molecular mechanisms of heparin/alginate-induced angiogenesis. The physical properties of the hydrogels were evaluated by Fourier transform infrared spectroscopy (FTIR). Results: FTIR confirmed the formation of heparin-loaded alginate wound dressing and the compatibility of both heparin and alginate. Among all, 10 µg/mL concentration of heparin showed the best antibacterial activity against E. coli. The swelling was considerably increased up to 1500% within 1 h. Alamar Blue assay revealed no cytotoxic effect on NIH3T3. Heparin showed good anti-microbial properties and inhibited the growth of E. coli in zones with a diameter of 18 mm. The expression analysis suggested that heparin probably exerts its pro-angiogenetic effect through VEGF and cPGE. Conclusions: We report that heparin-loaded alginate dressings are not cytotoxic and offer increased angiogenic and anti-bacterial potential. The angiogenesis is apparently taken through the VEGF pathway.
Collapse
Affiliation(s)
- Ayesha Nawaz
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
- Department of Biology, Lahore Garrison University, Lahore 54810, Pakistan
| | - Sher Zaman Safi
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom 42610, Selangor, Malaysia
| | - Shomaila Sikandar
- Department of Biology, Lahore Garrison University, Lahore 54810, Pakistan
| | - Rabia Zeeshan
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Saima Zulfiqar
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Nadia Mehmood
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Hussah M. Alobaid
- Department of Zoology, College of Science, King Saud University, Riyadh 11362, Saudi Arabia
| | - Fozia Rehman
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Muhammad Imran
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar 25120, Pakistan
| | - Muhammad Tariq
- Department of Medical Laboratory Technology, University College of Duba, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Abid Ali
- Department of Zoology, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Muhammad Yar
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| |
Collapse
|
187
|
Bu S, Nguyen HC, Nikfarjam S, Michels DCR, Rasheed B, Maheshkumar S, Singh S, Singh KK. Endothelial cell-specific loss of eNOS differentially affects endothelial function. PLoS One 2022; 17:e0274487. [PMID: 36149900 PMCID: PMC9506615 DOI: 10.1371/journal.pone.0274487] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/29/2022] [Indexed: 12/01/2022] Open
Abstract
The endothelium maintains and regulates vascular homeostasis mainly by balancing interplay between vasorelaxation and vasoconstriction via regulating Nitric Oxide (NO) availability. Endothelial nitric oxide synthase (eNOS) is one of three NOS isoforms that catalyses the synthesis of NO to regulate endothelial function. However, eNOS’s role in the regulation of endothelial function, such as cell proliferation and migration remain unclear. To gain a better understanding, we genetically knocked down eNOS in cultured endothelial cells using sieNOS and evaluated cell proliferation, migration and also tube forming potential in vitro. To our surprise, loss of eNOS significantly induced endothelial cell proliferation, which was associated with significant downregulation of both cell cycle inhibitor p21 and cell proliferation antigen Ki-67. Knockdown of eNOS induced cell migration but inhibited formation of tube-like structures in vitro. Mechanistically, loss of eNOS was associated with activation of MAPK/ERK and inhibition of PI3-K/AKT signaling pathway. On the contrary, pharmacologic inhibition of eNOS by inhibitors L-NAME or L-NMMA, inhibited cell proliferation. Genetic and pharmacologic inhibition of eNOS, both promoted endothelial cell migration but inhibited tube-forming potential. Our findings confirm that eNOS regulate endothelial function by inversely controlling endothelial cell proliferation and migration, and by directly regulating its tube-forming potential. Differential results obtained following pharmacologic versus genetic inhibition of eNOS indicates a more complex mechanism behind eNOS regulation and activity in endothelial cells, warranting further investigation.
Collapse
Affiliation(s)
- Shuhan Bu
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Hien C. Nguyen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sepideh Nikfarjam
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - David C. R. Michels
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Berk Rasheed
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sauraish Maheshkumar
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Shweta Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Krishna K. Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
188
|
AbouSamra MM, El Hoffy NM, El-Wakil NA, Awad GEA, Kamel R. Computational Investigation to Design Ofloxacin-Loaded Hybridized Nanocellulose/Lipid Nanogels for Accelerated Skin Repair. Gels 2022; 8:gels8090593. [PMID: 36135305 PMCID: PMC9498533 DOI: 10.3390/gels8090593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
The pharmaceutical application of biomaterials has attained a great success. Rapid wound healing is an important goal for many researchers. Hence, this work deals with the development of nanocellulose crystals/lipid nanogels loaded with ofloxacin (OFX) to promote skin repair while inhibiting bacterial infection. Ofloxacin-loaded hybridized nanocellulose/lipid nanogels (OFX-HNCNs) were prepared and evaluated adopting a computational method based on regression analysis. The optimized nanogels (OFX-HNCN7) showed a spherical outline with an encapsulation efficiency (EE), particle size (PS) and zeta potential (ZP) values of 97.53 ± 1.56%, 200.2 ± 6.74 nm and -26.4 ± 0.50 mV, respectively, with an extended drug release profile. DSC examination of OFX-HNCN7 proved the amorphization of the encapsulated drug into the prepared OFX-HNCNs. Microbiological studies showed the prolonged inhibition of bacterial growth by OFX-HNCN7 compared to the free drug. The cytocompatibility of OFX-HNCN7 was proved by Sulforhodamine B assay. Tissue repair was evaluated using the epidermal scratch assay based on cell migration in human skin fibroblast cell line, and the results depicted that cell treated with OFX-HNCN7 showed a faster and more efficient healing compared to the control. In overall, the obtained findings emphasize the benefits of using the eco-friendly bioactive nanocellulose, hybridized with lipid, to prepare a nanocarrier for skin repair.
Collapse
Affiliation(s)
- Mona M. AbouSamra
- Pharmaceutical Technology Department, National Research Centre, Giza 12622, Egypt
| | - Nada M. El Hoffy
- Faculty of Pharmacy, Future University in Egypt, New Cairo 11835, Egypt
- Correspondence: (N.M.E.H.); or (R.K.); Tel.: +20-100-80-20-20-2 (N.M.E.H.); +20-11-13-63-91-93 (R.K.)
| | - Nahla A. El-Wakil
- Cellulose and Paper Department, National Research Centre, Giza 12622, Egypt
| | - Ghada E. A. Awad
- Chemistry of Natural and Microbial Product Department, National Research Centre, Giza 12622, Egypt
| | - Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre, Giza 12622, Egypt
- Correspondence: (N.M.E.H.); or (R.K.); Tel.: +20-100-80-20-20-2 (N.M.E.H.); +20-11-13-63-91-93 (R.K.)
| |
Collapse
|
189
|
Liu N, Bowen CM, Shoja MM, Castro de Pereira KL, Dongur LP, Saad A, Russell WK, Broderick TC, Fair JH, Fagg WS. Comparative Analysis of Co-Cultured Amniotic Cell-Conditioned Media with Cell-Free Amniotic Fluid Reveals Differential Effects on Epithelial–Mesenchymal Transition and Myofibroblast Activation. Biomedicines 2022; 10:biomedicines10092189. [PMID: 36140291 PMCID: PMC9495976 DOI: 10.3390/biomedicines10092189] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Myofibroblast activation is a cellular response elicited by a variety of physiological or pathological insults whereby cells initiate a coordinated response intended to eradicate the insult and then revert back to a basal state. However, an underlying theme in various disease states is persistent myofibroblast activation that fails to resolve. Based on multiple observations, we hypothesized that the secreted factors harvested from co-culturing amniotic stem cells might mimic the anti-inflammatory state that cell-free amniotic fluid (AF) elicits. We optimized an amnion epithelial and amniotic fluid cell co-culture system, and tested this hypothesis in the context of myofibroblast activation. However, we discovered that co-cultured amniotic cell conditioned media (coACCM) and AF have opposing effects on myofibroblast activation: coACCM activates the epithelial–mesenchymal transition (EMT) and stimulates gene expression patterns associated with myofibroblast activation, while AF does the opposite. Intriguingly, extracellular vesicles (EVs) purified from AF are necessary and sufficient to activate EMT and inflammatory gene expression patterns, while the EV-depleted AF potently represses these responses. In summary, these data indicate that coACCM stimulates myofibroblast activation, while AF represses it. We interpret these findings to suggest that coACCM, AF, and fractionated AF represent unique biologics that elicit different cellular responses that are correlated with a wide variety of pathological states, and therefore could have broad utility in the clinic and the lab.
Collapse
Affiliation(s)
- Naiyou Liu
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Charles M. Bowen
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mohammadali M. Shoja
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | - Laxmi Priya Dongur
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Antonio Saad
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William K. Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Thomas Christopher Broderick
- Merakris Therapeutics, RTP Frontier, Research Triangle Park, NC 27709, USA
- Golden LEAF Biomanufacturing Training and Education Center, North Carolina State University, Raleigh, NC 27606, USA
| | - Jeffrey H. Fair
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William Samuel Fagg
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Merakris Therapeutics, RTP Frontier, Research Triangle Park, NC 27709, USA
- Correspondence: ; Tel.: +1-(409)-772-2412; Fax: +1-(409)-747-7364
| |
Collapse
|
190
|
Activation of non-classical NMDA receptors by glycine impairs barrier function of brain endothelial cells. Cell Mol Life Sci 2022; 79:479. [PMID: 35951110 PMCID: PMC9372018 DOI: 10.1007/s00018-022-04502-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/04/2022] [Accepted: 07/16/2022] [Indexed: 12/24/2022]
Abstract
Blood–brain barrier (BBB) integrity is necessary to maintain homeostasis of the central nervous system (CNS). NMDA receptor (NMDAR) function and expression have been implicated in BBB integrity. However, as evidenced in neuroinflammatory conditions, BBB disruption contributes to immune cell infiltration and propagation of inflammatory pathways. Currently, our understanding of the pathophysiological role of NMDAR signaling on endothelial cells remains incomplete. Thus, we investigated NMDAR function on primary mouse brain microvascular endothelial cells (MBMECs). We detected glycine-responsive NMDAR channels, composed of functional GluN1, GluN2A and GluN3A subunits. Importantly, application of glycine alone, but not glutamate, was sufficient to induce NMDAR-mediated currents and an increase in intracellular Ca2+ concentrations. Functionally, glycine-mediated NMDAR activation leads to loss of BBB integrity and changes in actin distribution. Treatment of oocytes that express NMDARs composed of different subunits, with GluN1 and GluN3A binding site inhibitors, resulted in abrogation of NMDAR signaling as measured by two-electrode voltage clamp (TEVC). This effect was only detected in the presence of the GluN2A subunits, suggesting the latter as prerequisite for pharmacological modulation of NMDARs on brain endothelial cells. Taken together, our findings argue for a novel role of glycine as NMDAR ligand on endothelial cells shaping BBB integrity.
Collapse
|
191
|
Suh YK, Robinson A, Zanghi N, Kratz A, Gustetic A, Crow MM, Ritts T, Hankey W, Segarra VA. Introducing Wound Healing Assays in the Undergraduate Biology Laboratory Using Ibidi Plates. JOURNAL OF MICROBIOLOGY & BIOLOGY EDUCATION 2022; 23:e00061-22. [PMID: 36061326 PMCID: PMC9429954 DOI: 10.1128/jmbe.00061-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/24/2022] [Indexed: 06/15/2023]
Abstract
The wound healing assay is a simple and inexpensive method that allows researchers to experimentally mimic cell growth and migration leading to wound healing. In this assay, a wound is created on a monolayer of cultured mammalian cells and cell migration is monitored. Micrographs are captured at regular intervals during the duration of the experiment. These microscopy images are analyzed to compare cell migration and wound closure under different conditions. Introduction of different cytotoxic treatments into a wound healing assay can provide information as to whether a particular drug or compound of interest has the ability to affect cell migration. This type of analysis is important when assessing the ability of a particular cancer cell line to display invasive and metastatic behaviors. One of the challenges of this assay is to create the original wound in a way that is consistent across plates or treatments, facilitating comparisons across experimental groups. This is a particular challenge when using the wound healing assay in the context of an undergraduate biology class to expose students to a distinct form of mammalian cell culture and help them apply scientific knowledge and research skills. We found an easy way to overcome this obstacle by using ibidi plates. In this article, we provide a simple protocol to use ibidi plates and HeLa cells to set up wound healing assays. This laboratory exercise allows undergraduate students to utilize different skills developed through cell culture experience, such as growing, treating, and imaging mammalian cells.
Collapse
Affiliation(s)
- Yewseok K. Suh
- Department of Biology, High Point University, High Point, North Carolina, USA
| | - Ashley Robinson
- Department of Biology, High Point University, High Point, North Carolina, USA
| | - Nicholas Zanghi
- Department of Biology, High Point University, High Point, North Carolina, USA
| | - Austin Kratz
- Department of Chemistry, High Point University, High Point, North Carolina, USA
- Department of Physics, High Point University, High Point, North Carolina, USA
| | - Andrew Gustetic
- Department of Biology, High Point University, High Point, North Carolina, USA
| | - Mackenzie M. Crow
- Department of Biology, High Point University, High Point, North Carolina, USA
| | - Taylor Ritts
- Department of Biology, High Point University, High Point, North Carolina, USA
| | - William Hankey
- Department of Genetics, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, USA
| | - Verónica A. Segarra
- Department of Biology, High Point University, High Point, North Carolina, USA
| |
Collapse
|
192
|
Vrazas V, Moustafa S, Makridakis M, Karakasiliotis I, Vlahou A, Mavromara P, Katsani KR. A Proteomic Approach to Study the Biological Role of Hepatitis C Virus Protein Core+1/ARFP. Viruses 2022; 14:v14081694. [PMID: 36016316 PMCID: PMC9518822 DOI: 10.3390/v14081694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatitis C virus is the major cause of chronic liver diseases and the only cytoplasmic RNA virus known to be oncogenic in humans. The viral genome gives rise to ten mature proteins and to additional proteins, which are the products of alternative translation initiation mechanisms. A protein-known as ARFP (alternative reading frame protein) or Core+1 protein-is synthesized by an open reading frame overlapping the HCV Core coding region in the (+1) frame of genotype 1a. Almost 20 years after its discovery, we still know little of the biological role of the ARFP/Core+1 protein. Here, our differential proteomic analysis of stable hepatoma cell lines expressing the Core+1/Long isoform of HCV-1a relates the expression of the Core+1/Long isoform with the progression of the pathology of HCV liver disease to cancer.
Collapse
Affiliation(s)
- Vasileios Vrazas
- Laboratory of Biochemistry and Molecular Virology, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (V.V.); (P.M.)
| | - Savvina Moustafa
- Clinical Immunology-Rheumatology Unit, 2nd Department of Medicine and Laboratory, Hippokration General Hospital of Athens, 11527 Athens, Greece;
| | - Manousos Makridakis
- Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (A.V.); (M.M.)
| | - Ioannis Karakasiliotis
- Laboratory of Biology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Antonia Vlahou
- Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (A.V.); (M.M.)
| | - Penelope Mavromara
- Laboratory of Biochemistry and Molecular Virology, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (V.V.); (P.M.)
| | - Katerina R. Katsani
- Laboratory of Biochemistry and Molecular Virology, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (V.V.); (P.M.)
- Correspondence:
| |
Collapse
|
193
|
Gonzalez-Salinas F, Martinez-Amador C, Trevino V. Characterizing genes associated with cancer using the CRISPR/Cas9 system: A systematic review of genes and methodological approaches. Gene 2022; 833:146595. [PMID: 35598687 DOI: 10.1016/j.gene.2022.146595] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/22/2022] [Accepted: 05/16/2022] [Indexed: 12/24/2022]
Abstract
The CRISPR/Cas9 system enables a versatile set of genomes editing and genetic-based disease modeling tools due to its high specificity, efficiency, and accessible design and implementation. In cancer, the CRISPR/Cas9 system has been used to characterize genes and explore different mechanisms implicated in tumorigenesis. Different experimental strategies have been proposed in recent years, showing dependency on various intrinsic factors such as cancer type, gene function, mutation type, and technical approaches such as cell line, Cas9 expression, and transfection options. However, the successful methodological approaches, genes, and other experimental factors have not been analyzed. We, therefore, initially considered more than 1,300 research articles related to CRISPR/Cas9 in cancer to finally examine more than 400 full-text research publications. We summarize findings regarding target genes, RNA guide designs, cloning, Cas9 delivery systems, cell enrichment, and experimental validations. This analysis provides valuable information and guidance for future cancer gene validation experiments.
Collapse
Affiliation(s)
- Fernando Gonzalez-Salinas
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Morones Prieto avenue 3000, Monterrey, Nuevo Leon 64710, Mexico
| | - Claudia Martinez-Amador
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Morones Prieto avenue 3000, Monterrey, Nuevo Leon 64710, Mexico
| | - Victor Trevino
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Morones Prieto avenue 3000, Monterrey, Nuevo Leon 64710, Mexico; Tecnologico de Monterrey, The Institute for Obesity Research, Eugenio Garza Sada avenue 2501, Monterrey, Nuevo Leon 64849, México.
| |
Collapse
|
194
|
Masitas C, Peng Z, Wang M, Konai MM, Avila-Cobian LF, Lemieux L, Hovanesian J, Grady JE, Mobashery S, Chang M. Matrix Metalloproteinase-14 as an Instigator of Fibrosis in Human Pterygium and Its Pharmacological Intervention. ACS Pharmacol Transl Sci 2022; 5:555-561. [DOI: 10.1021/acsptsci.2c00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Cesar Masitas
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Zhihong Peng
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Man Wang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mohini Mohan Konai
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Luis F. Avila-Cobian
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Leslie Lemieux
- Harvard Eye Associates, Laguna Hills, California 92653, United States
| | - John Hovanesian
- Harvard Eye Associates, Laguna Hills, California 92653, United States
| | - James E. Grady
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
195
|
Sohrabi Kashani A, Larocque K, Piekny A, Packirisamy M. Gold Nano-Bio-Interaction to Modulate Mechanobiological Responses for Cancer Therapy Applications. ACS APPLIED BIO MATERIALS 2022; 5:3741-3752. [PMID: 35839330 DOI: 10.1021/acsabm.2c00230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the present study, we investigate the mechanobiological responses of human lung cancer that may occur through their interactions with two different types of gold nanoparticles: nanostars and nanospheres. Hyperspectral images of nanoparticle-treated cells revealed different spatial distributions of nanoparticles in cells depending on their morphology, with nanospheres being more uniformly distributed in cells than nanostars. Gold nanospheres were also found to be more effective in mechanobiological modulations. They significantly suppressed the migratory ability of cells under different incubation times while lowering the bulk stiffness and adhesion of cells. This in vitro study suggests the potential applications of gold nanoparticles to manage cell migration. Nano-bio-interactions appeared to impact the cytoskeletal organization of cells and consequently alter the mechanical properties of cells, which could influence the cellular functions of cells. According to the results and migratory index model, it is thought that nanoparticle-treated cells experience mechanical changes in their body, which largely reduces their migratory potentials. These findings provide a better understanding of nano-bio-interaction in terms of cell mechanics and highlight the importance of mechanobiological responses in designing gold nanoparticles for cancer therapy.
Collapse
Affiliation(s)
- Ahmad Sohrabi Kashani
- Optical Bio-Microsystem Lab, Micro-Nano-Bio-Integration Centre, Department of Mechanical, Industrial and Aerospace Engineering of Concordia University, 1455 De Maisonneuve Blvd. W., Montreal, Quebec, Canada, H3G 1M8
| | - Kevin Larocque
- Department of Biology, Concordia University, 7141 Sherbrooke Street W., Montreal, Quebec, Canada, H4B 1R6
| | - Alisa Piekny
- Department of Biology, Concordia University, 7141 Sherbrooke Street W., Montreal, Quebec, Canada, H4B 1R6
| | - Muthukumaran Packirisamy
- Optical Bio-Microsystem Lab, Micro-Nano-Bio-Integration Centre, Department of Mechanical, Industrial and Aerospace Engineering of Concordia University, 1455 De Maisonneuve Blvd. W., Montreal, Quebec, Canada, H3G 1M8
| |
Collapse
|
196
|
Zhang H, Kim HT, Feeley BT, Lin G, Lue TF, Liu M, Banie L, Liu X. Microenergy acoustic pulses promotes muscle regeneration through in situ activation of muscle stem cells. J Orthop Res 2022; 40:1621-1631. [PMID: 34657315 PMCID: PMC9013392 DOI: 10.1002/jor.25184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/16/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023]
Abstract
Microenergy acoustic pulses (MAP) is a modified low-intensity extracorporeal shock wave therapy that currently used for treating musculoskeletal disorders. However, its function on muscle regeneration after ischemia-reperfusion injury (IRI) remains unknown. This study aimed to explore the effect of MAP on muscle injury after IRI and its underlying mechanisms. Ten-week-old C57BL/6J mice underwent unilateral hindlimb IRI followed with or without MAP treatment. Wet weight of tibialis anterior muscles at both injury and contralateral sides were measured followed with histology analysis at 3 weeks after IRI. In in vitro study, the myoblasts, endothelial cells and fibro-adipogenic progenitors (FAP) were treated with MAP. Cell proliferation and differentiation were assessed, and related gene expressions were measured by real-time PCR. Our results showed that MAP significantly increased the muscle weight and centrally nucleated regenerating muscle fiber size along with a trend in activating satellite cells. In vitro data indicated that MAP promoted myoblast proliferation and differentiation and endothelial cells migration. MAP also induced FAP brown/beige adipogenesis, a promyogenic phenotype of FAPs. Our findings demonstrate the beneficial function of MAP in promoting muscle regeneration after IR injury by inducing muscle stem cells proliferation and differentiation.
Collapse
Affiliation(s)
- He Zhang
- Department of Physical Education, Central South University, Hunan, China,Department of Orthopaedic Surgery, San Francisco Veterans Affair Health Care System, San Francisco, CA, USA,Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - Hubert T. Kim
- Department of Orthopaedic Surgery, San Francisco Veterans Affair Health Care System, San Francisco, CA, USA,Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - Brian T. Feeley
- Department of Orthopaedic Surgery, San Francisco Veterans Affair Health Care System, San Francisco, CA, USA,Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Tom F. Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Mengyao Liu
- Department of Orthopaedic Surgery, San Francisco Veterans Affair Health Care System, San Francisco, CA, USA,Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Xuhui Liu
- Department of Orthopaedic Surgery, San Francisco Veterans Affair Health Care System, San Francisco, CA, USA,Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, CA, USA,Corresponding author: Xuhui Liu, MD, 1700 Owens Street, San Francisco, CA 94158, Tel: 415-575-0546, Fax: 415-750-2181,
| |
Collapse
|
197
|
Li D, Ma H, Shu Q, Wang T, Li L, Huang P, Lou K, Xu H. Arsenite inhibits M2a polarization of macrophages through downregulation of peroxisome proliferator-activated receptor gamma. Toxicol Appl Pharmacol 2022; 450:116142. [PMID: 35777529 DOI: 10.1016/j.taap.2022.116142] [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: 03/20/2022] [Revised: 06/08/2022] [Accepted: 06/23/2022] [Indexed: 11/28/2022]
Abstract
Arsenite (As+3) is a group one human carcinogen, which has been associated with many diseases. Previous studies indicated that As+3 could inhibit wound healing and repair. M2a cells are known as tissue remodeling macrophages, which play an important role in wound repair process. Peroxisome proliferator-activated receptor gamma (PPAR-γ), a key regulator of lipid and glucose metabolism, was found to mediate the IL-4-dependent M2a polarization of macrophages. In the present study, As+3 induced dose-dependent inhibition of M2a polarization starting from 0.1 μM in THP-1-derived macrophages stimulated with 20 ng/mL IL-4. Increased lipid accumulation and decreased PPAR-γ expression were also observed in As+3-treated M2a macrophages. Rosiglitazone (RSG), a potent PPAR-γ agonist, alleviated the suppressions of PPAR-γ and M2a polarization induced by 2 μM As+3. Collectively, these results not only demonstrated that As+3 was able to inhibit polarization of M2a cells through PPAR-γ suppression, but also indicated that PPAR-γ could be utilized as a target for the prevention and treatment of As+3-induced immunotoxicity.
Collapse
Affiliation(s)
- Dan Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Huijuan Ma
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Qi Shu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Tingqian Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Linyi Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Ping Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Kaiyan Lou
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| | - Huan Xu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
198
|
Combined Curcumin and Luteolin Synergistically Inhibit Colon Cancer Associated with Notch1 and TGF-β Signaling Pathways in Cultured Cells and Xenograft Mice. Cancers (Basel) 2022; 14:cancers14123001. [PMID: 35740666 PMCID: PMC9221484 DOI: 10.3390/cancers14123001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/06/2022] [Accepted: 06/16/2022] [Indexed: 02/01/2023] Open
Abstract
This study aimed to select a combination of curcumin and luteolin, two phytochemicals from food, at lower concentrations with a higher inhibitory effect on colon cancer growth and investigate possible molecular mechanisms of this anti-colon cancer effect. By pairwise combination screening, we identified that the combination of curcumin (CUR) at 15 μM and luteolin (LUT) at 30 μM (C15L30) synergistically suppressed the proliferation of human colon cancer CL-188 cells, but the individual chemicals had a little inhibitory effect at the selected concentrations. This result was also confirmed in other colon cancer DLD-1cells, suggesting that this synergistic inhibitory effect of C15L30 applies to different colon cancer cells. The combination C15L30 synergistically suppressed the wound closure (wound healing assay) in CL-188 cells. We also found that the combination of CUR and LUT (at 20 mg/kg/day and 10 mg/kg/day, respectively, IP injection, 5 days for 2 weeks) synergistically suppressed tumor growth in CL-188 cell-derived xenograft mice. Western blot results showed that protein levels of Notch1 and TGF-β were synergistically reduced by the combination, both in CL-188 cells and xenograft tumors. Tumor pathological analysis revealed that combined CUR and LUT synergistically increased necrosis, but the individual treatment with CUR and LUT had no significant effect on tumor necrosis. Therefore, combined curcumin and luteolin synergically inhibit colon cancer development by suppressing cell proliferation, necrosis, and migration associated with Notch1 and TGF-β pathways. This study provides evidence that colon cancer may be prevented/treated by consuming foods having high levels of luteolin and curcumin in humans.
Collapse
|
199
|
Webb G. The force of cell-cell adhesion in determining the outcome in a nonlocal advection diffusion model of wound healing. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:8689-8704. [PMID: 35942731 DOI: 10.3934/mbe.2022403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A model of wound healing is presented to investigate the connection of the force of cell-cell adhesion to the sensing radius of cells in their spatial environment. The model consists of a partial differential equation with nonlocal advection and diffusion terms, describing the movement of cells in a spatial environment. The model is applied to biological wound healing experiments to understand incomplete wound closure. The analysis demonstrates that for each value of the force of adhesion parameter, there is a critical value of the sensing radius above which complete wound healing does not occur.
Collapse
Affiliation(s)
- Glenn Webb
- Mathematics Department, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
200
|
Boger KD, Sheridan AE, Ziegler AL, Blikslager AT. Mechanisms and modeling of wound repair in the intestinal epithelium. Tissue Barriers 2022; 11:2087454. [PMID: 35695206 PMCID: PMC10161961 DOI: 10.1080/21688370.2022.2087454] [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: 10/18/2022] Open
Abstract
The intestinal epithelial barrier is susceptible to injury from insults, such as ischemia or infectious disease. The epithelium's ability to repair wounded regions is critical to maintaining barrier integrity. Mechanisms of intestinal epithelial repair can be studied with models that recapitulate the in vivo environment. This review focuses on in vitro injury models and intestinal cell lines utilized in such systems. The formation of artificial wounds in a controlled environment allows for the exploration of reparative physiology in cell lines modeling diverse aspects of intestinal physiology. Specifically, the use of intestinal cell lines, IPEC-J2, Caco-2, T-84, HT-29, and IEC-6, to model intestinal epithelium is discussed. Understanding the unique systems available for creating intestinal injury and the differences in monolayers used for in vitro work is essential for designing studies that properly capture relevant physiology for the study of intestinal wound repair.
Collapse
Affiliation(s)
- Kasey D Boger
- Comparative Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Ana E Sheridan
- Comparative Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Amanda L Ziegler
- Comparative Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Anthony T Blikslager
- Comparative Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|