1
|
Jiang Y, He H, Jia X. Protective Role of Oxycodone in Myocardial Oxidative Stress and Mitochondrial Dysfunction Induced by Ischemia-Reperfusion. J Biochem Mol Toxicol 2025; 39:e70151. [PMID: 39865943 DOI: 10.1002/jbt.70151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
Ischemia-reperfusion (I/R) injury is a significant clinical problem impacting the heart and other organs, such as the kidneys and liver. This study explores the protective effects of oxycodone on myocardial I/R injury and its underlying mechanisms. Using a myocardial I/R model in Sprague-Dawley (SD) rats and an oxygen-glucose deprivation/reoxygenation (OGD/R) model in H9c2 cells, we administered oxycodone and inhibited AMP-activated protein kinase (AMPK) with Compound C (C.C). Our results showed that oxycodone significantly reduced lactate dehydrogenase (LDH) release and reactive oxygen species (ROS) production while stabilizing mitochondrial membrane potential (MMP). Western blot and RT-qPCR analyzes confirmed that oxycodone enhances AMPK phosphorylation and upregulates the expression of Silent Information Regulator 1 (SIRT1) and Peroxisome Proliferator-Activated Receptor γ Coactivator 1α (PGC-1α), thereby protecting myocardial cells. These findings suggest that oxycodone exerts significant protective effects against I/R injury by activating the AMPK pathway, offering new potential therapeutic targets for myocardial protection.
Collapse
Affiliation(s)
- Yongzheng Jiang
- The People's Hospital of Jiawang District of Xuzhou City, Xuzhou, China
| | - Hua He
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Xinwei Jia
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
2
|
Zhou H, Wu Z, Zhang Y, Yu Z, Nie Z, Fan J, Zhu Z, Chen F, Wang T. In vitro anticancer study of novel curcumin derivatives via targeting PI3K/Akt/p53 signaling pathway. Mol Divers 2025; 29:73-86. [PMID: 38951417 DOI: 10.1007/s11030-024-10833-9] [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: 12/20/2023] [Accepted: 02/28/2024] [Indexed: 07/03/2024]
Abstract
Four new series of curcumin derivatives bearing NO-donating moiety were synthesized via etherification, nucleophilic substitution, and Knoevenagel condensation etc. The cytotoxicity activity of curcumin derivatives against five human tumor cell lines (A549, Hela, HepG2, MCF-7 and HT-29) and two normal cell lines (LO-2 and HK-2) has been studied. The results showed that compound 6a could inhibit the proliferation of MCF-7 cells remarkably and exhibit low toxicity to normal cells. Also, the underlying mechanism in vitro of compound 6a on MCF-7 was investigated. It has been found that compound 6a induced G2/M arrest and apoptosis of MCF-7 in a dose-dependent manner. Compound 6a-induced the fluorescence changes of ROS in MCF-7 cells confirmed the occurrence of apoptosis. Western Blot suggested that compound 6a decreased the expression of PI3K, as well as increased the expression of p53, cleaved caspase-9 and cleaved caspase-3. Furthermore, molecular docking revealed that compound 6a could bind well at active site of PI3K (3zim) with total score 9.59. Together, compound 6a, a potential PI3K inhibitor, may inhibit the survival of MCF-7 cells via interfering with PI3K/Akt/p53 pathway.
Collapse
Affiliation(s)
- Huixian Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Zhiwen Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Yannan Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Zikai Yu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Zhengyang Nie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Jinbiao Fan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Zuchang Zhu
- Technological R&D department, Lizhu Pharmaceutical Co., Ltd, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Fenglian Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China.
| | - Tao Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China.
| |
Collapse
|
3
|
Chen L, Chen M, Xie Y, Zhang Y, Mo S, He Y, Liang T, Liao Y, Huang R, Huang G, Han C, Pham TTH. 2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione mediates the effect of ROS-enhanced PI3K/Akt/mTOR pathway on autophagy in breast cancer. FEBS Open Bio 2024. [PMID: 39648951 DOI: 10.1002/2211-5463.13940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 12/10/2024] Open
Abstract
Several studies have suggested a potential antitumor effect of 2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione (DMDD). To further understand the mechanism of action of this compound, we investigated its effect on the phosphatidylinositol-3-kinase (PI3K)/serine-threonine kinase (Akt)/mammalian target of rapamycin (mTOR) signaling pathway. We show that DMDD application significantly inhibited the proliferation of breast cancer cell lines MDA-MB-231 and ER-α positive MCF-7. Furthermore, DMDD application resulted in increased intracellular reactive oxygen species (ROS) levels, apoptosis and autophagy, whereas it downregulated the expression of PI3K, Akt and mTOR mRNA and proteins, and increased the expression of LC3II/I and p62 proteins. In a mouse breast cancer xenograft model, DMDD inhibited tumor growth. Expression analyses suggest that ROS levels were higher in DMDD treated tumor tissues, whereas immunohistochemical analyses suggest that apoptotic cells were more prevalent in the DMDD treated group compared to the control group. Taken together, our results suggest that the molecular mechanism of action of DMDD may involve the enhancement of breast cancer autophagy through the PI3K/Akt/mTOR signaling pathway by mediating ROS expression.
Collapse
Affiliation(s)
- Linqian Chen
- Guangxi Medical University School of Pharmacy, Nanning, China
| | - Meifeng Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yan Xie
- Guangxi Medical University School of Pharmacy, Nanning, China
| | - Yuyan Zhang
- Guilin Medical College School of Pharmacy, Guilin, China
| | - Shutian Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yongfei He
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tianyi Liang
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuan Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Renbin Huang
- Guangxi Medical University School of Pharmacy, Nanning, China
| | - Guodong Huang
- Zhuang & Yao Medicine Research and Development Center, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Thi Thai Hoa Pham
- Zhuang & Yao Medicine Research and Development Center, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
4
|
Ahmed NM, Mohamed MS, Awad SM, Abd El-Hameed RH, El-tawab NAA, Gaballah MS, Said AM. Design, synthesis, molecular modelling and biological evaluation of novel 6-amino-5-cyano-2-thiopyrimidine derivatives as potent anticancer agents against leukemia and apoptotic inducers. J Enzyme Inhib Med Chem 2024; 39:2304625. [PMID: 38348824 PMCID: PMC10866072 DOI: 10.1080/14756366.2024.2304625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
Herein, a novel series of 6-amino-5-cyano-2-thiopyrimidines and condensed pyrimidines analogues were prepared. All the synthesized compounds (1a-c, 2a-c, 3a-c, 4a-r and 5a-c) were evaluated for in vitro anticancer activity by the National Cancer Institute (NCI; MD, USA) against 60 cell lines. Compound 1c showed promising anticancer activity and was selected for the five-dose testing. Results demonstrated that compound 1c possessed broad spectrum anti-cancer activity against the nine cancerous subpanels tested with selectivity ratio ranging from 0.7 to 39 at the GI50 level with high selectivity towards leukaemia. Mechanistic studies showed that Compound 1c showed comparable activity to Duvelisib against PI3Kδ (IC50 = 0.0034 and 0.0025 μM, respectively) and arrested cell cycle at the S phase and displayed significant increase in the early and late apoptosis in HL60 and leukaemia SR cells. The necrosis percentage showed a significant increase from 1.13% to 3.41% in compound 1c treated HL60 cells as well as from 1.51% to 4.72% in compound 1c treated leukaemia SR cells. Also, compound 1c triggered apoptosis by activating caspase 3, Bax, P53 and suppressing Bcl2. Moreover, 1c revealed a good safety profile against human normal lung fibroblast cell line (WI-38 cells). Molecular analysis of Duvelisib and compound 1c in PI3K was performed. Finally, these results suggest that 2-thiopyrimidine derivative 1c might serve as a model for designing novel anticancer drugs in the future.
Collapse
Affiliation(s)
- Naglaa M. Ahmed
- Pharmaceutical Organic Chemistry Department, Helwan University, Ein-Helwan, Egypt
| | - Mosaad S. Mohamed
- Pharmaceutical Organic Chemistry Department, Helwan University, Ein-Helwan, Egypt
| | - Samir M. Awad
- Pharmaceutical Organic Chemistry Department, Helwan University, Ein-Helwan, Egypt
| | | | | | - Mohamed S. Gaballah
- Biochemistry and Molecular Biology Department, Helwan University, Ein-Helwan, Egypt
| | - Ahmed M. Said
- Pharmaceutical Organic Chemistry Department, Helwan University, Ein-Helwan, Egypt
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, USA
- Athenex Inc, Buffalo, NY, USA
| |
Collapse
|
5
|
Cherik IT, Divsalar A, Abdolhamid Angaji S. Design, Synthesis, and Toxicity Evaluation of the Green Synthesized Oxaliplatin Nanoparticles Using Ginger Extract against Colorectal Cancer Cells. IRANIAN JOURNAL OF SCIENCE 2024; 48:1411-1423. [DOI: 10.1007/s40995-024-01678-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/01/2024] [Indexed: 01/05/2025]
|
6
|
Majidzadeh H, Araj-Khodaei M, Aghanejad A, Ghaffari M, Jafari A, Jenanifard F, Ezzati Nazhad Dolatabadi J, Andishmand H, Hamblin MR. PAMAM dendrimers based co-delivery of methotrexate and berberine for targeting of Hela cancer cells. Toxicol Rep 2024; 13:101765. [PMID: 39484635 PMCID: PMC11525225 DOI: 10.1016/j.toxrep.2024.101765] [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: 07/31/2024] [Revised: 10/05/2024] [Accepted: 10/09/2024] [Indexed: 11/03/2024] Open
Abstract
Polyamidoamine dendrimer (PAMAM) is a class of synthetic macromolecular polymers for targeted drug delivery. PAMAM dendrimers are characterized by a pure defined structure, adjustable nanoscale dimensions, mono-dispersity, and versatile surface modification. The objective of this study was to study the covalent coupling of methotrexate (MTX) to PAMAM dendrimer, which was loaded with the natural product anticancer agent, berberine (BER) for the targeting of HeLa cells. The successful preparation of MTX-conjugated PAMAM loaded with BER (MTX-PAMAM-BER) was confirmed by Fourier transform infrared spectroscopy and particle size was evaluated using dynamic light scattering. The biological assays, MTT, flow cytometry, ROS levels evaluation and DAPI staining were used to assess the cytotoxicity effect of the prepared nanosystem. The findings indicated that MTX-PAMAM-BER exhibited greater suppression of tumor cell growth in comparison to BER, MTX, PAMAM-BER, and MTX-PAMAM. Besides, the noteworthy ROS level has been seen in the treated cells with MTX-PAMAM-BER. Finally, it should be stated that the fabricated MTX-PAMAM-BER co-delivery nanosystem could be a promising agent for cancer therapy and targeting.
Collapse
Affiliation(s)
- Hossein Majidzadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Persian Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Araj-Khodaei
- Department of Persian Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Ghaffari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Jafari
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Jenanifard
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hashem Andishmand
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| |
Collapse
|
7
|
Meng D, Yang S, Ju L, Wang J, Yang Y, Zhang L, Cui L. Cell membrane camouflaged Cu-doped mesoporous polydopamine for combined CT/PTT/CDT synergistic treatment of breast cancer. Biomed Pharmacother 2024; 180:117539. [PMID: 39383733 DOI: 10.1016/j.biopha.2024.117539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024] Open
Abstract
Currently, traditional monotherapy for cancer often results in indiscriminate attacks on the body, leading to the emergence of new health problems. To confront these challenges, multimodal combination therapy has become necessary. However, how to develop new smart nanomaterials through green synthesis methods, delivering drugs while simultaneously synergizing multimodal combination therapies for tumor treatment, remains a topic of great significance. In this study, a biomimetic composite nanomaterial (RM-Cu/P) composed of mesoporous polydopamine (MPDA) as the core and red blood cell membranes (RBCMs) as the shell was synthesized as a drug carrier to deliver doxorubicin (DOX) while achieving synergistic chemotherapy, photothermal and chemodynamic therapy (CT/PTT/CDT). Herein, the nanoparticles were extensively characterized to examine their morphological characteristics, elemental composition, and drug-carrying capacity. Notably, the coating of RBCM reduced the toxicity of the RM-Cu/P@DOX nanoparticles, improved their targeting ability and prolonged their circulation time in vivo. The Cu-doped nanoparticles were capable of initiating a Fenton-like reaction to generate reactive oxygen species (ROS) for CDT, while the photothermal conversion efficiency (η) reached 45.20 % under NIR laser irradiation. Subsequently, the particles were examined by in vivo and in vitro experimental studies in cytotoxicity, cellular uptake, ROS levels, lysosomal escape, and mouse tumor model to evaluate their potential application in antitumor. Compared with monotherapy, the RM-Cu/P@DOX nanoparticles had multiple-stimulation response properties under redox, pH, and NIR, which exhibited the advantage of combined trimodal therapy, resulting in remarkable synergistic antitumor efficacy. In conclusion, this innovative platform exhibited promising applications in smart drug delivery and synergistic treatment of cancer.
Collapse
Affiliation(s)
- Di Meng
- School of Biological Engineering, Henan University of Technology, Zhengzhou, PR China
| | - Shuoye Yang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, PR China.
| | - Lin Ju
- School of Biological Engineering, Henan University of Technology, Zhengzhou, PR China
| | - Jinpeng Wang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, PR China
| | - Yanan Yang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, PR China
| | - Lu Zhang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, PR China
| | - Lan Cui
- School of Biological Engineering, Henan University of Technology, Zhengzhou, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, PR China
| |
Collapse
|
8
|
Mahmoud Z, Ismail MM, Kamel M, Youssef A. Levofloxacin reposition-based design: synthesis, biological evaluation of new levofloxacin derivatives targeting topoisomerase II beta polymerase as promising anticancer agents, molecular docking, and physicochemical characterization. RSC Adv 2024; 14:28098-28119. [PMID: 39228758 PMCID: PMC11369887 DOI: 10.1039/d4ra03975k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/12/2024] [Indexed: 09/05/2024] Open
Abstract
Repositioning of already approved medications through repurposing or re-profiling for new medical uses after certain structural modifications is a novel approach in drug discovery. Fluoroquinolone antibiotics are one of the cardinal agents investigated for potential anticancer activities. In this work, levofloxacin was repositioned for anticancer activities. A series of levofloxacin-based compounds were designed and synthesized through the derivatization of levofloxacin's carboxylic acid functionality. The newly synthesized compounds were screened for cytotoxic activities against breast, liver, and leukemia cancer cell lines. Their effect on normal cells was also investigated. The target compounds were evaluated for their proliferative inhibitory activity toward topoisomerase II beta polymerization. Compound 5 showed higher inhibitory activity against a breast cancer cell line (MCF-7) with IC50 = 1.4 μM and lesser side effects on a normal breast cell line (MCF-10a) with IC50 = 30.40 μM than reference drugs. The best activity against a liver cancer cell line (Hep3B) was exhibited by compounds 3c, 4b, 5, 7, 8, 13a and 13c with IC50 values ranging from 0.43 to 8.79 μM. Regarding the effect of compounds 5 and 13a on a leukemia cancer cell line (L-SR), their IC50 values were 0.96 and 3.12 μM, respectively. Compounds 3c and 5 showed Topo2β inhibitory effects on Hep3B cells (81.33% and 83.73%, respectively), which was better than levofloxacin and etoposide as reference drugs. Cytometry cell cycle analysis revealed that compounds 3c and 5 arrested the cell cycle at the S phase (37.56% and 39.09%, respectively). Compounds 3c and 5 exhibited an elevation in active caspase-3 levels by 4.9 and 4.5 folds, respectively. Molecular modeling simulation of compounds 3c and 5 demonstrated energy scores (-29.77 and -20.46 kcal mol-1, respectively) more than those of the reference drugs as they interact with the most essential amino acids required for good affinity towards human topoisomerase II beta enzyme (PDB ID: 3QX3). Physicochemical characteristics of the most promising cytotoxic compounds 3c and 5 were investigated and compared to etoposide and levofloxacin as reference drugs. However, they showed high gastrointestinal absorption and could not penetrate the blood-brain barrier.
Collapse
Affiliation(s)
- Zeinab Mahmoud
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University 11561 Cairo Egypt
| | - Mohamed M Ismail
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology P. O. Box 77 Giza Egypt +201285266644
| | - Mona Kamel
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University 11561 Cairo Egypt
| | - Amira Youssef
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology P. O. Box 77 Giza Egypt +201285266644
| |
Collapse
|
9
|
D’Antonio DL, Marchetti S, Pignatelli P, Umme S, De Bellis D, Lanuti P, Piattelli A, Curia MC. Effect of 5-Aminolevulinic Acid (5-ALA) in "ALADENT" Gel Formulation and Photodynamic Therapy (PDT) against Human Oral and Pancreatic Cancers. Biomedicines 2024; 12:1316. [PMID: 38927525 PMCID: PMC11201195 DOI: 10.3390/biomedicines12061316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Oral squamous-cell and pancreatic carcinomas are aggressive cancers with a poor outcome. Photodynamic therapy (PDT) consists of the use of photosensitizer-induced cell and tissue damage that is activated by exposure to visible light. PDT selectively acts on cancer cells, which have an accumulation of photosensitizer superior to that of the normal surrounding tissues. 5-aminolevulinic acid (5-ALA) induces the production of protoporphyrin IX (PpIX), an endogenous photosensitizer activated in PDT. This study aimed to test the effect of a new gel containing 5% v/v 5-ALA (ALAD-PDT) on human oral CAL-27 and pancreatic CAPAN-2 cancer cell lines. The cell lines were incubated in low concentrations of ALAD-PDT (0.05%, 0.10%, 0.20%, 0.40%, 0.75%, 1.0%) for 4 h or 8 h, and then irradiated for 7 min with 630 nm RED light. The cytotoxic effects of ALAD-PDT were measured using the MTS assay. Apoptosis, cell cycle, and ROS assays were performed using flow cytometry. PpIX accumulation was measured using a spectrofluorometer after 10 min and 24 and 48 h of treatment. The viability was extremely reduced at all concentrations, at 4 h for CAPAN-2 and at 8 h for CAL-27. ALAD-PDT induced marked apoptosis rates in both oral and pancreatic cancer cells. Elevated ROS production and appreciable levels of PpIX were detected in both cell lines. The use of ALA-PDT as a topical or intralesional therapy would permit the use of very low doses to achieve effective results and minimize side effects. ALAD-PDT has the potential to play a significant role in complex oral and pancreatic anticancer therapies.
Collapse
Affiliation(s)
- Domenica Lucia D’Antonio
- Department of Medical, Oral and Biotechnological Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.L.D.); (S.M.); (S.U.)
- Villa Serena Foundation for Research, Via Leonardo Petruzzi 42, 65013 Città Sant’Angelo, Italy
| | - Simona Marchetti
- Department of Medical, Oral and Biotechnological Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.L.D.); (S.M.); (S.U.)
| | - Pamela Pignatelli
- COMDINAV DUE, Nave Cavour, Italian Navy, Stazione Navale Mar Grande, Viale Jonio, 74122 Taranto, Italy;
| | - Samia Umme
- Department of Medical, Oral and Biotechnological Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.L.D.); (S.M.); (S.U.)
| | - Domenico De Bellis
- Center for Advanced Studies and Technology (CAST), “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.D.B.); (P.L.)
- Department of Medicine and Aging Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Paola Lanuti
- Center for Advanced Studies and Technology (CAST), “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.D.B.); (P.L.)
- Department of Medicine and Aging Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Adriano Piattelli
- School of Dentistry, Saint Camillus International University of Health and Medical Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy;
- Facultad de Medicina, UCAM Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
| | - Maria Cristina Curia
- Department of Medical, Oral and Biotechnological Sciences, “Gabriele d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (D.L.D.); (S.M.); (S.U.)
| |
Collapse
|
10
|
Kęska A, Rusak A, Włostowski R, Dziemieszkiewicz M, Szymlet N. Low-vacuum SEM imaging and viability test of L929 cells exposed to a Euro 6 diesel exhaust gas mixture in a BAT-CELL chamber in comparison with hydrocarbons emission. Sci Rep 2024; 14:12883. [PMID: 38839874 PMCID: PMC11153568 DOI: 10.1038/s41598-024-63560-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024] Open
Abstract
Exhaust emissions, which count among the most common causes of premature death worldwide, can cause irreversible changes in cells, leading to their damage or degeneration. In this research, L929 line cells were observed after exposure in the BAT-CELL chamber to exhaust gases emitted from a Euro 6 compression-ignition engine. Real road traffic conditions were simulated, taking into account air resistance while driving at speeds of 50 km/h, 120 km/h and idling engine. Morphological analysis of the cells was performed using an environmental scanning electron microscope. It has been observed that diesel exhaust fumes can cause inflammation, which can induce apoptosis or leads to necrotic cell death. The impact of the vehicle exhaust gases can inhibit cell proliferation by almost three times. Moreover, a correlation has been observed between the speed of the inflammatory reaction in cells and the presence of specific hydrocarbon compounds that determine the toxicity of exhaust gases. Research has shown that the toxicity of the emitted exhaust gases has been the highest at the driving speed of 120 km/h. In order to reduce the harmful effects of exhaust emissions, ecological alternatives and the supplementation of legal provisions regarding the compounds subject to limitation are necessary.
Collapse
Affiliation(s)
- Aleksandra Kęska
- Department of Automotive Engineering, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland.
| | - Agnieszka Rusak
- Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chałubińskiego 6a, 50-368, Wrocław, Poland
| | - Radosław Włostowski
- Department of Automotive Engineering, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland
| | | | - Natalia Szymlet
- Institute of Combustion Engines and Powertrains, Poznan University of Technology, Pl. Marii Skłodowskiej-Curie 5, 60-965, Poznań, Poland
| |
Collapse
|
11
|
Wang H, Zhang L, Huang J, Yang Z, Fan C, Yuan L, Zhao H, Zhang Z, Liu X. Imaging the intracellular refractive index distribution (IRID) for dynamic label-free living colon cancer cells via circularly depolarization decay model (CDDM). BIOMEDICAL OPTICS EXPRESS 2024; 15:2451-2465. [PMID: 38633098 PMCID: PMC11019712 DOI: 10.1364/boe.518957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 04/19/2024]
Abstract
Label-free detection of intracellular substances for living cancer cells remains a significant hurdle in cancer pathogenesis research. Although the sensitivity of light polarization to intracellular substances has been validated, current studies are predominantly focused on tissue lesions, thus label-free detection of substances within individual living cancer cells is still a challenge. The main difficulty is to find specific detection methods along with corresponding characteristic parameters. With refractive index as an endogenous marker of substances, this study proposes a detection method of intracellular refractive index distribution (IRID) for label-free living colon cancer (LoVo) cells. Utilizing the circular depolarization decay model (CDDM) to calculate the degree of circular polarization (DOCP) modulated by the cell allows for the derivation of the IRID on the focal plane. Experiments on LoVo cells demonstrated the refractive index of single cell can be accurately and precisely measured, with precision of 10-3 refractive index units (RIU). Additionally, chromatin content during the interphases (G1, S, G2) of cell cycle was recorded at 56.5%, 64.4%, and 71.5%, respectively. A significantly finer IRID can be obtained compared to the phase measurement method. This method is promising in providing a dynamic label-free intracellular substances detection method in cancer pathogenesis studies.
Collapse
Affiliation(s)
- Huijun Wang
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Lu Zhang
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- School of Instrument Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Jie Huang
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Zewen Yang
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Chen Fan
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- School of Instrument Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Li Yuan
- First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| | - Hong Zhao
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- School of Instrument Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Zhenxi Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi’an Jiaotong University, Xi’an 710049, China
| | - Xiaolong Liu
- Mengchao Hepatobiliary Hospital of Fujian Medical University, The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Provincey, Fuzhou 350025, China
| |
Collapse
|
12
|
Khalef L, Lydia R, Filicia K, Moussa B. Cell viability and cytotoxicity assays: Biochemical elements and cellular compartments. Cell Biochem Funct 2024; 42:e4007. [PMID: 38593323 DOI: 10.1002/cbf.4007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/01/2024] [Accepted: 03/29/2024] [Indexed: 04/11/2024]
Abstract
Cell viability and cytotoxicity assays play a crucial role in drug screening and evaluating the cytotoxic effects of various chemicals. The quantification of cell viability and proliferation serves as the cornerstone for numerous in vitro assays that assess cellular responses to external factors. In the last decade, several studies have developed guidelines for defining and interpreting cell viability and cytotoxicity based on morphological, biochemical, and functional perspectives. As this domain continues to experience ongoing growth, revealing new mechanisms orchestrating diverse cell cytotoxicity pathways, we suggest a revised classification for multiple assays employed in evaluating cell viability and cell death. This classification is rooted in the cellular compartment and/or biochemical element involved, with a specific focus on mechanistic and essential aspects of the process. The assays are founded on diverse cell functions, encompassing metabolic activity, enzyme activity, cell membrane permeability and integrity, adenosine 5'-triphosphate content, cell adherence, reduction equivalents, dye inclusion or exclusion, constitutive protease activity, colony formation, DNA fragmentation and nuclear splitting. These assays present straightforward, reliable, sensitive, reproducible, cost-effective, and high-throughput approaches for appraising the effects of newly formulated chemotherapeutic biomolecules on the cell survival during the drug development process.
Collapse
Affiliation(s)
- Lefsih Khalef
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| | - Radja Lydia
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| | - Khettar Filicia
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| | - Berkoud Moussa
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| |
Collapse
|
13
|
Rady GS, El Deeb MA, Sarg MTM, Taher AT, Helwa AA. Design, synthesis and biological evaluation of novel morpholinopyrimidine-5-carbonitrile derivatives as dual PI3K/mTOR inhibitors. RSC Med Chem 2024; 15:733-752. [PMID: 38389871 PMCID: PMC10880895 DOI: 10.1039/d3md00693j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 12/17/2023] [Indexed: 02/24/2024] Open
Abstract
In this study, novel morpholinopyrimidine-5-carbonitriles were designed and synthesized as dual PI3K/mTOR inhibitors and apoptosis inducers. The integration of a heterocycle at position 2, with or without spacers, of the new key intermediate 2-hydrazinyl-6-morpholinopyrimidine-5-carbonitrile (5) yielded compounds 6-10, 11a-c and 12a-h. The National Cancer Institute (USA) tested all compounds for antiproliferative activity. Schiff bases, 12a-h analogs, were the most active ones. The most promising compounds 12b and 12d exhibited excellent antitumor activity against the leukemia SR cell line, which is the most sensitive cell line, with IC50 0.10 ± 0.01 and 0.09 ± 0.01 μM, respectively, along with significant effects on PI3Kα/PI3Kβ/PI3Kδ with IC50 values of 0.17 ± 0.01, 0.13 ± 0.01 and 0.76 ± 0.04 μM, respectively, for 12b and 1.27 ± 0.07, 3.20 ± 0.16 and 1.98 ± 0.11, respectively, for 12d compared to LY294002. Compared to Afinitor, these compounds inhibited mTOR with IC50 values of 0.83 ± 0.05 and 2.85 ± 0.17 μM, respectively. Annexin-V and propidium iodide (PI) double labeling showed that compounds 12b and 12d promote cytotoxic leukemia SR apoptosis. Compounds 12b and 12d also caused a G2/M cell cycle arrest in the leukaemia SR cell line. The findings of this study indicate that the highest effect was observed for 12b, which was supported by western blot and docking analysis.
Collapse
Affiliation(s)
- Ghada S Rady
- Directorate of Health Affairs in Giza, Ministry of Health Egypt
| | - Moshira A El Deeb
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University Cairo Egypt
| | - Marwa T M Sarg
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University Cairo Egypt
| | - Azza T Taher
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University Cairo 11562 Egypt
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, October 6 University (O6U) 6th of October city Giza 12585 Egypt
| | - Amira A Helwa
- Department of Pharmaceutical Organic Chemistry, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST) Al-Motamayez District, P.O. Box: 77, 6th of October city Giza Egypt
| |
Collapse
|
14
|
Wang M, Feng Y, Zhang P, Shen K, Su J, Zhong Y, Yang X, Lin S, Lu J. Jiawei Maxing Shigan Tang alleviates radiation-induced lung injury via TGF-β1/Smad signaling pathway mediated by regulatory T cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117389. [PMID: 37944875 DOI: 10.1016/j.jep.2023.117389] [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: 09/03/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radiation-induced lung injury (RILI) is a common complication during thoracic radiotherapy which impairs the quality of life in patients and limits radiation doses. Jiawei Maxing Shigan Tang (JMST), which is a modified decoction made of Ephedra, Apricot Kernel, Gypsum, and Licorice, can alleviate the symptoms of RILI in patients. Previous studies and preliminary findings suggested a potential molecular mechanism of JMST in the treatment of RILI. Further studies are needed. AIM OF THE STUDY To elucidate the mechanisms of how regulatory T cells (Tregs) promote RILI and the effect of JMST on Tregs, as well as the corresponding pathway. MATERIALS AND METHODS CD4+CD25+ Tregs were isolated from rats, and the supernatant's TGF-β1 level was examined by using enzyme-linked immunosorbent assay (ELISA). Type II alveolar epithelial cells (AECs) were co-cultured with the supernatant of Tregs, and the expression levels of epithelial-to-mesenchymal transition (EMT)-related and TGF-β1/Smad signaling pathway-related proteins were analyzed by using western blotting (WB). Afterward, the Tregs were incubated with different concentrations of JMST. The cell viability and TGF-β1 concentration were confirmed by cell counting kit-8 (CCK-8) assay and ELISA, respectively. The optimized concentration of JMST was applied in vitro and vivo experiments. The specific mechanism was investigated through the combination of using flow cytometry, lung histopathology analysis, ELISA, and WB. RESULTS Radiation could promote Tregs to secrete TGF-β1. After radiation, the expression levels of Smad2/3, phosphorylated Smad2/3 (p-Smad2/3), Smad4 and mesenchymal markers Vimentin and α-SMA were all increased, while the expression level of epithelial markers E-cadherin was decreased. The expression levels of these proteins were reversed after interventions involving Treg cell activation inhibition or TGF-β1 receptor inhibitor. JMST reduced the number of Tregs in lung tissue and alleviated the degree of pulmonary fibrosis. The expression of Smad2/3, p-Smad2/3, Smad4, TGF-β1, Vimentin, and α-SMA were significantly downregulated, while the E-cadherin was upregulated, through the intervention of JMST. CONCLUSION Tregs could mediate EMT through TGF-β1/Smad pathway. JMST inhibits EMT via TGF-β1/Smad pathway by regulating Tregs, therefore alleviating RILI.
Collapse
Affiliation(s)
- Menglei Wang
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuqian Feng
- Hangzhou School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Pengcheng Zhang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kezhan Shen
- Hangzhou School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingyang Su
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yazhen Zhong
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Xuefei Yang
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China.
| | - Shengyou Lin
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Province Hospital of Chinese Medicine), Hangzhou, China.
| | - Jinhua Lu
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China.
| |
Collapse
|
15
|
Lazarski CA, Hanley PJ. Review of flow cytometry as a tool for cell and gene therapy. Cytotherapy 2024; 26:103-112. [PMID: 37943204 PMCID: PMC10872958 DOI: 10.1016/j.jcyt.2023.10.005] [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: 08/16/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Quality control testing and analytics are critical for the development and manufacture of cell and gene therapies, and flow cytometry is a key quality control and analytical assay that is used extensively. However, the technical scope of characterization assays and safety assays must keep apace as the breadth of cell therapy products continues to expand beyond hematopoietic stem cell products into producing novel adoptive immune therapies and gene therapy products. Flow cytometry services are uniquely positioned to support the evolving needs of cell therapy facilities, as access to flow cytometers, new antibody clones and improved fluorochrome reagents becomes more egalitarian. This report will outline the features, logistics, limitations and the current state of flow cytometry within the context of cellular therapy.
Collapse
Affiliation(s)
- Christopher A Lazarski
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| | - Patrick J Hanley
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| |
Collapse
|
16
|
Bhatnagar A, Mishra A. Development of Daruharidra ( Berberis aristata) Based Biogenic Cadmium Sulfide Nanoparticles: Their Implementation as Antibacterial and Novel Therapeutic Agents against Human Breast and Ovarian Cancer. Curr Pharm Biotechnol 2024; 25:1617-1628. [PMID: 39034838 DOI: 10.2174/0113892010244977231108043554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/18/2023] [Accepted: 10/03/2023] [Indexed: 07/23/2024]
Abstract
BACKGROUND This article presents a new and environmentally friendly method for generating DH-CdSNPs (cadmium sulfide nanoparticles) ranging from 5-10 nm in size. A green synthesis method for the development of inorganic nanoparticles was developed a few years back for their applications in diverse fields, such as medicine, bioimaging and remediation. The biogenic synthesis of these nanoparticles containing daruharidra (Berberis aristata) and cadmium sulfide is an effective alternative. AIMS By employing Daruharidra extract as a herbal analog, we aim to minimize the risks and adverse effects that come along with the use of other chemically synthesized nanoparticles. This study's main goal was to investigate the potential of these nanoparticles as powerful antibacterial and anticancer agents. METHODS We used a crude powdered daruharidra extract as a stabilizer ingredient to create CdSbased nanoformulations in an environmentally responsible way. By exposing the breast cancer cell line (MDAMB-231) and ovarian teratocarcinoma cell line (PA1) to these nanoformulations, we were able to evaluate their anticancer activities. Additionally, flow cytometry analysis was conducted to scrutinize the process of cell cycle arrest and apoptosis in reference to anticancer studies. Furthermore, DH-CdSNPs were applied on different gram-positive as well as gramnegative bacteria in a disc diffusion assay to ascertain their antibacterial activity. Nanoparticles were tested on bacterial strains to check if they were resistant after the MIC or minimum inhibitory concentration. RESULTS The cytotoxicity of nanoparticles was tested by MTT assay. The impact of increasing concentrations of NPs on cell lines was tested, revealing a cytotoxic effect. The half-maximal inhibitory concentration values for a 24-hour treatment were determined to be 95.74μg/ml for ovarian cancer cells and 796.25 μg/ml for breast cancer cells. Treatment with DH-CdSNP resulted in a noteworthy increase in early apoptotic cells, with percentages rising from approximately 3% to 14.5% in ovarian cancer cell lines and from 4% to 13.6% in breast cancer cell lines. Furthermore, the NPs induced arrest of the cell cycle, specifically in the interphase of G2 and mitosis phase, with DNA damage observed in sub G1 in ovarian cancer cells and G0/G1 arrest observed in breast cancer cells. Additionally, the NPs exhibited exceptional potency against both gram-positive as well as gram-negative bacteria. CONCLUSION Less research has been done on using bioinspired DH-CdSNP to deliver anticancer medications. The amalgamation of plant extract and the DH-CdSNP could cause a paradigm shift in the cancer therapy approach. The findings revealed that the biosynthesized DH-CdSNP limited the growth of human breast and ovarian cancer cells. This property can be further investigated against a variety of additional cell lines to determine whether this property makes the DH-CdSNP a promising treatment alternative. The results obtained from these nanoformulations exhibit faster efficacy compared to traditional medications.
Collapse
Affiliation(s)
- Aditi Bhatnagar
- School of Biochemical Engineering, IIT (BHU)-Varanasi-221005, India
| | - Abha Mishra
- School of Biochemical Engineering, IIT (BHU)-Varanasi-221005, India
| |
Collapse
|
17
|
Miedes D, Cilla A, Alegría A. Chemopreventive Effect of an In Vitro Digested and Fermented Plant Sterol-Enriched Wholemeal Rye Bread in Colon Cancer Cells. Foods 2023; 13:112. [PMID: 38201138 PMCID: PMC10778687 DOI: 10.3390/foods13010112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Diet is crucial for the prevention of colorectal cancer. Whole grains are the source of beneficial compounds for this, such as fiber. The enrichment of wholemeal rye bread with plant sterols (PSs) could increase its beneficial effects. This study aimed to assess the potential antiproliferative effect of this enriched food on colon adenocarcinoma cells (Caco-2) compared with a non-enriched one. After a human oral chewing, simulated semi-dynamic gastrointestinal digestion and colonic fermentation in a simgi® system, fermentation liquids (FLs) obtained were used as treatment for cells. Cytotoxicity assay showed that samples diluted 1/5 (v/v) with DMEM are not toxic for non-tumoral cells, whereas they damage tumoral cells. Samples with PS (FLPS) produced a higher chemopreventive effect (vs. blank) in MTT and apoptosis assays, as well as higher gene expression of TP53 and Casp8. Nevertheless, FL0 (without PS) produced a higher chemopreventive effect in a cell cycle and reduced glutathione and calcium assays, besides producing higher gene expression of Casp3 and lower CCND1. The distinct antiproliferative effect of both FLs is attributed to differences in PSs, short chain fatty acids (lower concentration in FLPS vs. FL0) and antioxidant compounds. These results may support wholemeal rye bread consumption as a way of reducing the risk of colorectal cancer development, although further research would be needed.
Collapse
Affiliation(s)
| | - Antonio Cilla
- Nutrition and Food Science Area, Faculty of Pharmacy, University of Valencia, Av. Vicente Andrés Estellés s/n, 46100 Burjassot, Spain; (D.M.); (A.A.)
| | | |
Collapse
|
18
|
Yao Y, Cai X, Zhang M, Zheng Y, Fei W, Zhao M, Zheng C. Circ-Plod2 destabilizes Mpo mRNA by binding to IGF2BP2 to promote osteogenic differentiation of bone marrow mesenchymal stem cells. Eur J Pharmacol 2023; 961:176192. [PMID: 37981258 DOI: 10.1016/j.ejphar.2023.176192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/11/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Abstract
Osteogenic differentiation, proliferation, and/or apoptosis of bone marrow mesenchymal stem cells (BMSCs) are involved in the progression of postmenopausal osteoporosis (PMO). However, circular RNA (circRNA)-mediated changes in the cellular function of BMSCs in PMO are still unclear. This study revealed the excellent ability of circ-Plod2 to promote osteogenic differentiation of BMSCs and its molecular mechanisms. In this study, ovariectomized (OVX) rats and control (Sham) rats were used to simulate PMO. Initially, we found that the expression of circ-Plod2 in OVX BMSCs is down-regulated and the expression of the Mpo gene is up-regulated by sequencing and verification. Further, we confirmed that circ-Plod2 is located in the cytoplasm and belongs to exon-type circRNA. Interestingly, circ-Plod2 promotes Mpo-dependent osteogenic differentiation of BMSCs without affecting proliferation, apoptosis, adipogenic differentiation, or chondrogenic differentiation of BMSCs. Mechanistically, we demonstrated that circ-Plod2 specifically binds IGF2BP2 to form an RNA-protein complex that destabilizes Mpo mRNA. Overexpression of circ-Plod2 in the bone marrow cavity effectively alleviated osteoporosis in OVX rats and inhibited the expression of MPO in BMSCs. Together, this study reveals that circ-Plod2 destabilizes Mpo mRNA by binding to IGF2BP2 to promote osteogenic differentiation of BMSCs to alleviate osteoporosis. The findings of this study may provide biomarkers for the diagnosis of PMO, and may also provide potential strategies for the clinical treatment of PMO.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xiaoyu Cai
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yongquan Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Mengdan Zhao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
19
|
Makran M, Garcia-Llatas G, Alegría A, Cilla A. Ethylcoprostanol modulates colorectal cancer cell proliferation and mitigates cytotoxicity of cholesterol metabolites in non-tumor colon cells. Food Funct 2023; 14:10829-10840. [PMID: 37982821 DOI: 10.1039/d3fo01868g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Sterols can be metabolized by gut microbiota. The cholesterol metabolites have been proposed as promoters of colorectal cancer (CRC), while the effect of plant sterol metabolites is unknown. This study aimed to evaluate the cytotoxicity of metabolites from cholesterol (coprostanol, cholestanol, coprostanone and cholestenone) and β-sitosterol (ethylcoprostanol) on human colon tumor (Caco-2) and non-tumor (CCD-18Co) cells at physiological concentrations (9-300 μM) and exposure time (24 h). Ethylcoprostanol reduced the tumor cell proliferation (MTT), showing in flow cytometry assays induction of apoptosis via production of reactive oxygen species (ROS) and ceramide. Transcriptomic analysis (qPCR) showed activation of the intrinsic apoptosis pathway (BAX/BCL2 ratio and CASP9 increased), accompanied by downregulation of the p21 gene. Cholesterol metabolites, mainly the most hydrophobic, induced apoptosis and G0/G1 phase arrest in non-tumor cells through overproduction of ROS. Both the intrinsic and extrinsic (CASP8 increased) apoptosis pathways occurred. In turn, a reduction in the expression of the cyclin E1 gene confirmed the cell cycle arrest. In addition, ethylcoprostanol protected non-tumor cells from the most cytotoxic cholesterol metabolite (cholestenone). In conclusion, ethylcoprostanol is a promising candidate as a therapeutic adjuvant in CRC, while cholesterol metabolites could act as CRC promoters through their cytotoxicity.
Collapse
Affiliation(s)
- Mussa Makran
- Nutrition and Food Science Area, Faculty of Pharmacy and Food Sciences, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain.
| | - Guadalupe Garcia-Llatas
- Nutrition and Food Science Area, Faculty of Pharmacy and Food Sciences, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain.
| | - Amparo Alegría
- Nutrition and Food Science Area, Faculty of Pharmacy and Food Sciences, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain.
| | - Antonio Cilla
- Nutrition and Food Science Area, Faculty of Pharmacy and Food Sciences, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain.
| |
Collapse
|
20
|
Fakhry MM, Mattar AA, Alsulaimany M, Al-Olayan EM, Al-Rashood ST, Abdel-Aziz HA. New Thiazolyl-Pyrazoline Derivatives as Potential Dual EGFR/HER2 Inhibitors: Design, Synthesis, Anticancer Activity Evaluation and In Silico Study. Molecules 2023; 28:7455. [PMID: 37959874 PMCID: PMC10647861 DOI: 10.3390/molecules28217455] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
A new series of thiazolyl-pyrazoline derivatives (4a-d, 5a-d 6a, b, 7a-d, 8a, b, and 10a, b) have been designed and synthesized through the combination of thiazole and pyrazoline moieties, starting from the key building blocks pyrazoline carbothioamides (1a-b). These eighteen derivatives have been designed as anticipated EGFR/HER2 dual inhibitors. The efficacy of the developed compounds in inhibiting cell proliferation was assessed using the breast cancer MCF-7 cell line. Among the new synthesized thiazolyl-pyrazolines, compounds 6a, 6b, 10a, and 10b displayed potent anticancer activity toward MCF-7 with IC50 = 4.08, 5.64, 3.37, and 3.54 µM, respectively, when compared with lapatinib (IC50 = 5.88 µM). In addition, enzymatic assays were also run for the most cytotoxic compounds (6a and 6b) toward EGFR and HER2 to demonstrate their dual inhibitory activity. They revealed promising inhibition potency against EGFR with IC50 = 0.024, and 0.005 µM, respectively, whereas their IC50 = 0.047 and 0.022 µM toward HER2, respectively, compared with lapatinib (IC50 = 0.007 and 0.018 µM). Both compounds 6a and 10a induced apoptosis by arresting the cell cycle of the MCF-7 cell line at the G1 and G1/S phases, respectively. Molecular modeling studies for the promising candidates 6a and 10a showed that they formed the essential binding with the crucial amino acids for EGFR and HER2 inhibition, supporting the in vitro assay results. Furthermore, ADMET study predictions were carried out for the compounds in the study.
Collapse
Affiliation(s)
- Mariam M. Fakhry
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Badr 11829, Egypt;
| | - Amr A. Mattar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Badr 11829, Egypt;
| | - Marwa Alsulaimany
- Department of Pharmacognosy & Pharmaceutical Chemistry, College of Pharmacy, Taibah University, Medina 42353, Saudi Arabia;
| | - Ebtesam M. Al-Olayan
- Department of Zoology, College of Science, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Sara T. Al-Rashood
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Hatem A. Abdel-Aziz
- Applied Organic Chemistry Department, National Research Center, Cairo 12622, Egypt
| |
Collapse
|
21
|
Reda N, Elshewy A, El-Askary HI, Mohamed KO, Helwa AA. Design, synthesis, and biological evaluation of new pyrimidine-5-carbonitrile derivatives as novel anti-cancer, dual EGFR WT/COX-2 inhibitors with docking studies. RSC Adv 2023; 13:32296-32320. [PMID: 37928843 PMCID: PMC10620772 DOI: 10.1039/d3ra06088h] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/20/2023] [Indexed: 11/07/2023] Open
Abstract
A novel series of pyrimidine-5-carbonitrile derivatives was designed, synthesized, then evaluated for their cytotoxic activity as novel anti-cancer with dual EGFRWT/COX-2 inhibitors. Two compounds 4e and 4f disclosed the highest activity against all NCI60 panel cell lines. They were most potent against Colo 205 (IC50 = 1.66, and 1.83 μM), Sequentially. The most potent two compounds disturbed cell cycle of Colo-205 cells by blocking the G1 phase, coupled with increased annexin-Vstained cells which indicated the increasing in percentage of apoptosis. In addition, 4e and 4f increase the concentration of caspase-3 by 10, and 8-fold compared to control, respectively. Moreover, the two candidate compounds were screened for cytotoxicity on normal epithelial colon cells; fortunately, they were found to be safe. Molecular docking study displayed that these compounds bound to the active site as EGFRWT/COX-2 inhibitors. Furthermore, 3D pharmacophore mapping disclosed many shared features between the most potent candidates 4e and 4f and the standard EGFRWT/COX-2 inhibitors; erlotinib, and celecoxib, respectively. Finally, the physicochemical parameter was calculated for the most potent novel anticancer candidates and the SwissAdme parameter showed that the newly synthesized compounds have good drug-likeness properties.
Collapse
Affiliation(s)
- Nada Reda
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST) 6th of October City Egypt
| | - Ahmed Elshewy
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt
- Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University New Galala 43713 Egypt
| | - Hesham I El-Askary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt
| | - Khaled O Mohamed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy. Sinai University (Arish Branch) El Arish Egypt
| | - Amira A Helwa
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST) 6th of October City Egypt
| |
Collapse
|
22
|
Zhu H, Liang H, Gao Z, Zhang X, He Q, He C, Cai C, Chen J. MiR-483-5p downregulation alleviates ox-LDL induced endothelial cell injury in atherosclerosis. BMC Cardiovasc Disord 2023; 23:521. [PMID: 37891465 PMCID: PMC10612234 DOI: 10.1186/s12872-023-03496-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/04/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND In light of the abnormal expression of microRNA (miR-483-5p) in patients with atherosclerosis (AS), its role in vascular endothelial cell injury was explored. And the mechanisms related to autophagy were also elucidated. METHODS Human umbilical vein endothelial cells (HUVECs) were given 100 mg/L ox-LDL to induce endothelial injury. Cell transfection was done to regulate miR-483-5p levels. Cell viability and apoptosis were detected. qRT-PCR was employed for the mRNA levels' detection. RESULTS Autophagic flux impairment of HUVECs was detected after ox-LDL treatment, along with the upregulation of miR-483-5p. Ox-LDL inhibited cell viability and promoted cell apoptosis, but these influences were changed by miR-483-5p downregulation. MiR-483-5p downregulation decreased the mRNA levels of IL-1β, IL-6, ICAM-1 and VCAM-1. 3-MA, the autophagy inhibitor, reversed the beneficial role of miR-483-5p downregulation in ox-LDL-induced HUVECs' injury. TIMP2 acts as a target gene of miR-483-5p, and was downregulated in HUVEC models. CONCLUSION MiR-483-5p downregulation alleviated ox-LDL-induced endothelial injury via activating autophagy, this might be related to TIMP2.
Collapse
Affiliation(s)
- Hezhong Zhu
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Hui Liang
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Zhen Gao
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, No. 32 Renminnan Road, Shiyan, 442000, China
| | - Xiaoqiao Zhang
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Qian He
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, No. 32 Renminnan Road, Shiyan, 442000, China
| | - Chaoyong He
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, No. 32 Renminnan Road, Shiyan, 442000, China
| | - Chao Cai
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, No. 32 Renminnan Road, Shiyan, 442000, China.
| | - Jiajuan Chen
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, No. 32 Renminnan Road, Shiyan, 442000, China.
| |
Collapse
|
23
|
Frutos-Grilo E, Kreling V, Hensel A, Campoy S. Host-pathogen interaction: Enterobacter cloacae exerts different adhesion and invasion capacities against different host cell types. PLoS One 2023; 18:e0289334. [PMID: 37874837 PMCID: PMC10597508 DOI: 10.1371/journal.pone.0289334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/17/2023] [Indexed: 10/26/2023] Open
Abstract
New antibiotics are urgently needed due to the huge increase of multidrug-resistant bacteria. The underexplored gram-negative bacterium Enterobacter cloacae is known to cause severe urinary tract and lung infections (UTIs). The pathogenicity of E. cloacae in UTI has only been studied at the bioinformatic level, but until now not within systematic in vitro investigations. The present study assesses different human cell lines for monitoring the early steps of host-pathogen interaction regarding bacterial adhesion to and invasion into different host cells by flow cytometric adhesion assay, classical cell counting assay, gentamicin invasion assay, and confocal laser scanning microscopy. To our knowledge, this is the first report in which E. cloacae has been investigated for its interaction with human bladder, kidney, skin, and lung cell lines under in vitro conditions. Data indicate that E. cloacae exerts strong adhesion to urinary tract (bladder and kidney) and lung cells, a finding which correlates with the clinical relevance of the bacterium for induction of urinary tract and lung infections. Furthermore, E. cloacae ATCC 13047 barely adheres to skin cells (A-431) and shows no relevant interaction with intestinal cells (Caco-2, HT-29), even in the presence of mucin (HT29 MTX). In contrast, invasion assays and confocal laser scanning microscopy demonstrate that E. cloacae internalizes in all tested host cells, but to a different extent. Especially, bladder and kidney cells are being invaded to the highest extent. Defective mutants of fimH and fimA abolished the adhesion of E. cloacae to T24 cells, while csgA deletion had no influence on adhesion. These results indicate that E. cloacae has different pattern for adhesion and invasion depending on the target tissue, which again correlates with the clinical relevance of the pathogen. For detailed investigation of the early host-pathogen interaction T24 bladder cells comprise a suitable assay system for evaluation the bacterial adhesion and invasion.
Collapse
Affiliation(s)
- Elisabet Frutos-Grilo
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Vanessa Kreling
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Andreas Hensel
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Susana Campoy
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
24
|
Li J, Fu R, Guo X, Pan Z, Xie J. Acupuncture improves immunity and fatigue after chemotherapy in breast cancer patients by inhibiting the Leptin/AMPK signaling pathway. Support Care Cancer 2023; 31:506. [PMID: 37542585 PMCID: PMC10404187 DOI: 10.1007/s00520-023-07967-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/24/2023] [Indexed: 08/07/2023]
Abstract
OBJECTIVE Acupuncture has become a popular complementary treatment in oncology. This study is based on RNA-Seq transcriptome sequencing technology to investigate the molecular mechanisms underlying the effect of acupuncture-mediated regulation of the Leptin/AMPK signaling pathway on mitochondrial dysfunction-induced fatigue in breast cancer patients after chemotherapy. METHODS Peripheral blood samples from 10 patients with post-operative chemotherapy for breast cancer were selected for transcriptome sequencing to screen the key molecular pathways involved in fatigue after chemotherapy in breast cancer patients. Besides, peripheral blood samples were collected from 138 post-operative chemotherapy patients with breast cancer to study the composite fatigue and quality of life scores. Flow cytometry was used to detect T lymphocyte subsets in peripheral blood-specific immune cells. In addition, a blood cell analyzer was used to measure peripheral blood leukocyte counts, and MSP-PCR was used to detect mitochondrial DNA mutations in peripheral blood leukocytes. RESULTS Transcriptome bioinformatics analysis screened 147 up-regulated mRNAs and 160 down-regulated mRNAs. Leptin protein was confirmed as the key factor. Leptin was significantly higher in the peripheral blood of breast cancer patients who developed fatigue after chemotherapy. Acupuncture treatment effectively improved post-chemotherapy fatigue and immune status in breast cancer patients, suppressed the expression of Leptin/AMPK signaling pathway-related factor and leukocyte counts, and significantly reduced the rate of mitochondrial DNA mutations in peripheral blood leukocytes. CONCLUSION The Leptin/AMPK signaling pathway may be the key molecular pathway affecting the occurrence of fatigue after chemotherapy in breast cancer patients. Leptin may improve post-chemotherapy fatigue in breast cancer patients by activating AMPK phosphorylation and alleviating mitochondrial functional impairment.
Collapse
Affiliation(s)
- Jinxia Li
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Ruiyang Fu
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Xiaoqing Guo
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Zhongqiang Pan
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Jingjun Xie
- Department of Rehabilitation Medicine, The First People's Hospital of Huzhou, No. 158, Guangchang Hou Road, Huzhou, 313000, Zhejiang, People's Republic of China.
| |
Collapse
|
25
|
Khosravi F, Mirzaei S, Hojati V, Hashemi M, Entezari M. Co-Administration of Vitamins B12 and D During Pregnancy Have Strong Neuroprotective Effects in Parkinson Disease. Mol Neurobiol 2023; 60:1986-1996. [PMID: 36596968 DOI: 10.1007/s12035-022-03186-7] [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: 09/02/2022] [Accepted: 12/21/2022] [Indexed: 01/05/2023]
Abstract
Parkinson's disease (PD) is a common disease whose pathophysiological mechanism is not well understood. Recent research studies have shown that PD patients have low serum levels of vitamins B12 and D. Therefore, in this study, the effects of supplementation with vitamins B12 and D on PD female mice as well as their fetuses were studied. After preparation of female mice and induction of Parkinson's by rotenone administration for 19 days, rotarod test was used to confirm PD induction. During this time, supplementations with vitamins B12 and D were performed. On day 19, after confirmation of PD induction, half of the mice were killed and the other half were allowed to mate with males. Viability was measured by the MTT method, and apoptosis and necrosis of cerebellar neurons were measured by flow cytometry. The RT-PCR technique was used to evaluate the relative expressions of the bax, bcl-2, miR-211, and circRNA 0,001,518 genes. Data analysis was performed by the GraphPad Prism V.8 software. Co-administration of vitamins B12 and D resulted in highest viability percentage and greatest reduction in apoptosis and necrosis of cerebellar neurons in the female mice as well as their fetuses compared to the PD females. A decrease in the relative expression of the bax and miR-211 genes and an increase in bcl-2 expression were observed in the cerebellar tissue of PD mice receiving both vitamins. Vitamins B12 and D have neuroprotective effects on PD conditions. Therefore, co-administration of these two vitamins is recommended in PD patients during pregnancy.
Collapse
Affiliation(s)
- Faramarz Khosravi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Vida Hojati
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran. .,Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran. .,Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
26
|
Punchoo R, Dreyer G, Pillay TS. 25-Hydroxycholecalciferol Inhibits Cell Growth and Induces Apoptosis in SiHa Cervical Cells via Autocrine Vitamin D Metabolism. Biomedicines 2023; 11:biomedicines11030871. [PMID: 36979850 PMCID: PMC10045786 DOI: 10.3390/biomedicines11030871] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Preclinical studies show that the anticancer actions of vitamin D metabolites are mediated by apoptosis, inhibition of cell proliferation and induction of cell cycle arrest. Cervical cancer cells express an autocrine vitamin D metabolising system (VDMS) comprised of a vitamin D receptor, vitamin D catabolic enzyme (CYP24A1), and the activating enzyme of 25-hydroxycholecalciferol (25(OH)D3), CYP27B1. We assessed the anticancer effects of 25(OH)D3 at clinically relevant concentrations on a cervical squamous cell cancer cell line, SiHa. We evaluated cell health parameters (cell count, viability, and cell cycle), cell death modes (apoptosis, autophagic-dependent death, and necrosis by flow cytometry and transmission electron microscopy), and autocrine VDMS gene and protein expression by qPCR and Western blot, respectively. Our study demonstrates that physiological and supraphysiological doses of 25(OH)D3 inhibit cell growth and viability and induce biochemical and morphological apoptosis in SiHa cells. These growth effects are mediated by alteration in the VDMS gene and protein expression, with prominent negative feedback at supraphysiological treatment dose. These data identify promising therapeutic potential of 25(OH)D3 in cervical cancer, which warrants further clinical translational investigations.
Collapse
Affiliation(s)
- Rivak Punchoo
- Tshwane Academic Division, National Health Laboratory Service (NHLS), Pretoria 0001, South Africa
- Department of Chemical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
- Correspondence: ; Tel.: +27-12-3192671
| | - Greta Dreyer
- Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Tahir S. Pillay
- Tshwane Academic Division, National Health Laboratory Service (NHLS), Pretoria 0001, South Africa
- Department of Chemical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
27
|
Emam SH, Hassan RA, Osman EO, Hamed MIA, Abdou AM, Kandil MM, Elbaz EM, Mikhail DS. Coumarin derivatives with potential anticancer and antibacterial activity: Design, synthesis, VEGFR-2 and DNA gyrase inhibition, and in silico studies. Drug Dev Res 2023; 84:433-457. [PMID: 36779381 DOI: 10.1002/ddr.22037] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 01/05/2023] [Accepted: 01/13/2023] [Indexed: 02/14/2023]
Abstract
A series of coumarin derivatives were designed, synthesized, and evaluated for their antiproliferative activity. Compound 3e exhibited significant antiproliferative activity and was further evaluated at five doses at the National Cancer Institute. It effectively inhibited vascular endothelial growth factor receptor-2 (VEGFR-2) with an IC50 value of 0.082 ± 0.004 µM compared with sorafenib. While compound 3e significantly downregulated total VEGFR-2 and its phosphorylation, it markedly reduced the HUVEC's migratory potential, resulting in a significant disruption in wound healing. Furthermore, compound 3e caused a 22.51-fold increment in total apoptotic level in leukemia cell line HL-60(TB) and a 6.91-fold increase in the caspase-3 level. Compound 3e also caused cell cycle arrest, mostly at the G1/S phase. Antibacterial activity was evaluated against Gram-positive and Gram-negative bacterial strains. Compound 3b was the most active derivative, with the same minimum inhibitory concentration and minimum bactericidal concentration value of 128 μg/mL against K. pneumonia and high stability in mammalian plasma. Moreover, compounds 3b and 3f inhibited Gram-negative DNA gyrase with IC50 = 0.73 ± 0.05 and 1.13 ± 0.07 µM, respectively, compared to novobiocin with an IC50 value of 0.17 ± 0.02 µM. The binding affinity and pattern of derivative 3e toward the VEGFR-2 active site and compounds 3a-c and 3f in the DNA gyrase active site were evaluated using molecular modeling. Overall, ADME studies of the synthesized coumarin derivatives displayed promising pharmacokinetic properties.
Collapse
Affiliation(s)
- Soha H Emam
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rasha A Hassan
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Eman O Osman
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohammed I A Hamed
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Amr M Abdou
- Department of Microbiology and Immunology, National Research Centre, Dokki, Giza, Egypt
| | - Mai M Kandil
- Department of Microbiology and Immunology, National Research Centre, Dokki, Giza, Egypt
| | - Eman Maher Elbaz
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Demiana S Mikhail
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
28
|
Huang YF, Ou GC, Ma SH, Liu MW, Deng W. Effect of icariin on the H 2O 2-induced proliferation of mouse airway smooth muscle cells through miR-138-5p regulating SIRT1/AMPK/PGC-1α axis. Int J Immunopathol Pharmacol 2023; 37:3946320231151515. [PMID: 36772811 PMCID: PMC9926010 DOI: 10.1177/03946320231151515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Abstract
Icariin exerts antioxidative and anti-inflammatory effects and is used in the treatment of bronchial asthma. However, the specific modes of action are uncertain. In this study, we investigated whether icariin could modulate the silencing information regulator 2-related enzyme 1 (SIRT1)/adenosine monophosphate-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor gamma co-activator 1α (PGC-1α) axis by regulating miR-138-5p during H2O2-induced proliferation of mouse airway smooth muscle cells (ASMCs). Primary BALB/c mouse ASMCs were cultured using the tissue block adherence method and were induced with hydrogen peroxide (H2O2; 200 μmol/L) to establish a bronchial asthma ASMC proliferation model. With the aid of Western Blot and quantitative real-time polymerase chain reaction (qRT-PCR) in H2O2-induced ASMCs, the expression of miR-138-5p, SIRT1, AMPK, PGC-1α, α-smooth muscle actin (α-SMA), transforming growth factor-β1 (TGF-β1), collagen I, and collagen III protein and mRNA were investigated. The proliferation rate and activities of superoxide dismutase1 (SOD1), reduced glutathione (GSH), malonaldehyde (MDA), and reactive oxygen species (ROS) in ASMCs were determined. The results suggest Compared with the H2O2-induced group, icariin inhibited the miR-138-5p expression; enhanced SIRT1, p-AMPK, and PGC-1α expression; attenuated MDA activity and ROS level; lowered TGF-β1, collagen I, and collagen III expression levels; and decreased the proliferation of ASMCs induced by H2O2. The dual-luciferase reporter gene assay results showed that SIRT1 is a regulatory target of miR-138-5p.The results suggest that Icariin could improve the H2O2-induced proliferation of ASMCs. The mechanism may be related to the increase of activation of SIRT1/AMPK/PGC-1α axis by suppressing the expression of miR-138-5p. Thus, SIRT1 is the regulatory target of miR-138-5p.
Collapse
Affiliation(s)
- Yu-fang Huang
- Department of Respiratory and
Critical Care, Suining
Central Hospital, Suining, China
| | - Guo-chun Ou
- Department of Respiratory and
Critical Care, Suining
Central Hospital, Suining, China
| | - Shou-hong Ma
- Medical Services Division,
Sixth
Affiliated Hospital of Kunming Medical
University, Yuxi, China
| | - Ming-wei Liu
- Department of Emergency,
First
Affiliated Hospital of Kunming Medical
University, Kunming, China,Ming-wei Liu, Department of Emergency,
First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wu
Hua District, Kunming 650051, China.
| | - Wen Deng
- Department of Emergency,
Suining
Central Hospital, Suining, China,Ming-wei Liu, Department of Emergency,
First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wu
Hua District, Kunming 650051, China.
| |
Collapse
|
29
|
Puri S, Stefan K, Khan SL, Pahnke J, Stefan SM, Juvale K. Indole Derivatives as New Structural Class of Potent and Antiproliferative Inhibitors of Monocarboxylate Transporter 1 (MCT1; SLC16A1). J Med Chem 2023; 66:657-676. [PMID: 36584238 PMCID: PMC9841531 DOI: 10.1021/acs.jmedchem.2c01612] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Indexed: 12/31/2022]
Abstract
The solute carrier (SLC) monocarboxylate transporter 1 (MCT1; SLC16A1) represents a promising target for the treatment of cancer; however, the MCT1 modulator landscape is underexplored with only roughly 100 reported compounds. To expand the knowledge about MCT1 modulation, we synthesized a library of 16 indole-based molecules and subjected these to a comprehensive biological assessment platform. All compounds showed functional inhibitory activities against MCT1 at low nanomolar concentrations and great antiproliferative activities against the MCT1-expressing cancer cell lines A-549 and MCF-7, while the compounds were selective over MCT4 (SLC16A4). Lead compound 24 demonstrated a greater potency than the reference compound, and molecular docking revealed strong binding affinities to MCT1. Compound 24 led to cancer cell cycle arrest as well as apoptosis, and it showed to sensitize these cancer cells toward an antineoplastic agent. Strikingly, compound 24 had also significant inhibitory power against the multidrug transporter ABCB1 and showed to reverse ABCB1-mediated multidrug resistance (MDR).
Collapse
Affiliation(s)
- Sachin Puri
- Shobhaben
Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s
NMIMS, V.L. Mehta Road,
Vile Parle (W), Mumbai400056, India
| | - Katja Stefan
- Department
of Pathology, Section of Neuropathology, Translational Neurodegeneration
Research and Neuropathology Lab (www.pahnkelab.eu), University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372Oslo, Norway
| | - Sharuk L. Khan
- Department
of Pharmaceutical Chemistry, N.B.S. Institute
of Pharmacy, Ausa413520, Maharashtra, India
| | - Jens Pahnke
- Department
of Pathology, Section of Neuropathology, Translational Neurodegeneration
Research and Neuropathology Lab (www.pahnkelab.eu), University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372Oslo, Norway
- Drug
Development and Chemical Biology Lab, Lübeck Institute of Experimental
Dermatology (LIED), University of Lübeck
and University Medical Center Schleswig-Holstein, Ratzeburger Allee 160, 23538Lübeck, Germany
- Department
of Pharmacology, Faculty of Medicine, University
of Latvia, Jelgavas iela
4, 1004Ri̅ga, Latvia
| | - Sven Marcel Stefan
- Department
of Pathology, Section of Neuropathology, Translational Neurodegeneration
Research and Neuropathology Lab (www.pahnkelab.eu), University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372Oslo, Norway
- Drug
Development and Chemical Biology Lab, Lübeck Institute of Experimental
Dermatology (LIED), University of Lübeck
and University Medical Center Schleswig-Holstein, Ratzeburger Allee 160, 23538Lübeck, Germany
| | - Kapil Juvale
- Shobhaben
Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s
NMIMS, V.L. Mehta Road,
Vile Parle (W), Mumbai400056, India
| |
Collapse
|
30
|
Challenging breast cancer through novel sulfonamide-pyridine hybrids: design, synthesis, carbonic anhydrase IX inhibition and induction of apoptosis. Future Med Chem 2023; 15:147-166. [PMID: 36762576 DOI: 10.4155/fmc-2022-0197] [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: 02/11/2023] Open
Abstract
Background: Among the important key modulators of the tumor microenvironment and hypoxia is a family of enzymes named carbonic anhydrases. Herein, 11 novel sulfonamide-pyridine hybrids (2-12) were designed, synthesized and biologically evaluated for their potential use in targeting breast cancer. Methods & results: The para chloro derivative 7 reported the highest cytotoxic activity against the three breast cancer cell lines used. In addition, compound 7 was found to induce cell cycle arrest and autophagy as well as delaying wound healing. The IC50 of compound 7 against carbonic anhydrase IX was 253 ± 12 nM using dorzolamide HCl as control. Conclusion: This study encourages us to expand the designed library, where more sulfonamide derivatives would be synthesized and studied for their structure-activity relationships.
Collapse
|
31
|
Zhao MH, Liu W, Zhang X, Zhang Y, Luo B. Epstein-Barr virus miR-BART2-5p and miR-BART11-5p regulate cell proliferation, apoptosis, and migration by targeting RB and p21 in gastric carcinoma. J Med Virol 2023; 95:e28338. [PMID: 36418188 DOI: 10.1002/jmv.28338] [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: 07/02/2022] [Revised: 10/12/2022] [Accepted: 11/12/2022] [Indexed: 11/25/2022]
Abstract
Epstein-Barr virus (EBV) was the first tumor virus discovered in humans and can cause various types of tumors. Molecular classification suggests that EBV-associated gastric cancer (EBVaGC) is a unique subtype of gastric cancer.EBV was also the first virus found to encode its own microRNAs. However, the functions of many miRNAs remain unknown. This study investigated the roles and targets of miR-BART2-5p (BART2-5p) and miR-BART11-5p (BART11-5p) in EBVaGC. The expression of RB and p21 in EBVaGC and EBV negative GC (EBVnGC) cells was evaluated by western blotting. Expression of BART2-5p and BART11-5p in EBVaGC cells was evaluated by droplet digital PCR. The effects of BART2-5p or BART11-5p and their potential mechanisms were further investigated using cell counting kit-8, colony formation assay, flow cytometry analysis, and transwell assay. BART2-5p and BART11-5p were abundantly expressed and RB and p21 were downregulated in EBVaGC cells. BART2-5p regulates RB and p21 expression by directly targeting them. BART11-5p regulates RB expression by directly targeting RB. Both BART2-5p and BART11-5p promoted proliferation and migration of gastric cancer cells, while inhibiting apoptosis and promoting S-phase arrest of the cell cycle. Thus, BART2-5p and BART11-5p play important roles in promoting proliferation and migration, and inhibiting apoptosis in EBVaGC by targeting RB and p21, thus providing new potential therapeutic targets for EBVaGC.
Collapse
Affiliation(s)
- Meng-He Zhao
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wen Liu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xing Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yan Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China.,Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Bing Luo
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
32
|
Coumaric acid from M. polymorphum extracts reverses the activated state of hepatic stellate cells (GRX) and inhibits their proliferation by decreasing the p53/p21 pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 396:925-937. [PMID: 36520165 DOI: 10.1007/s00210-022-02361-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
Coumaric acid is a phenolic compound found in medicinal plants. Its use has been reported in the treatment of inflammatory diseases, prevention of alterations induced by oxidative stress, as well as acetaminophen-induced hepatotoxicity. Thus, this study evaluated coumaric acid as a potential treatment for liver fibrosis. Cell proliferation was assessed by the trypan blue exclusion technique and the cytotoxicity of coumaric acid was performed using an LDH assay. Mechanisms of cell apoptosis were evaluated by flow cytometry. The expression of genes associated with apoptosis, cell cycle control, and fibrosis was assessed by qPCR. The production of lipid droplets was quantified by oil red staining. The experiments performed showed that the treatment with coumaric acid was able to reduce cell proliferation without causing cell cytotoxicity or apoptosis. Coumaric acid was able to inhibit the expression of cyclin D1 and CDK's (CDK2, CDK4, and CDK6), increasing p53 and p21, which could lead to cell cycle arrest. Treatment with coumaric acid was also able to revert the activated phenotype of GRX cells to their quiescent state. Thus, our results suggest that coumaric acid has a potential therapeutic effect against liver fibrosis.
Collapse
|
33
|
Dróżdż A, Sławińska-Brych A, Kubera D, Kimsa-Dudek M, Gola JM, Adamska J, Kruszniewska-Rajs C, Matwijczuk A, Karcz D, Dąbrowski W, Stepulak A, Gagoś M. Effect of Antibiotic Amphotericin B Combinations with Selected 1,3,4-Thiadiazole Derivatives on RPTECs in an In Vitro Model. Int J Mol Sci 2022; 23:ijms232315260. [PMID: 36499589 PMCID: PMC9738598 DOI: 10.3390/ijms232315260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
4-(5-methyl-1,3,4-thiadiazole-2-yl) benzene-1,3-diol (C1) and 4-[5-(naphthalen-1-ylmethyl)-1,3,4-thiadiazol-2-yl] benzene1,3-diol (NTBD) are representative derivatives of the thiadiazole group, with a high antimycotic potential and minimal toxicity against normal human fibroblast cells. The present study has proved its ability to synergize with the antifungal activity of AmB. The aim of this work was to evaluate the cytotoxic effects of C1 or NTBD, alone or in combination with AmB, on human renal proximal tubule epithelial cells (RPTECs) in vitro. Cell viability was assessed with the MTT assay. Flow cytometry and spectrofluorimetric techniques were used to assess the type of cell death and production of reactive oxygen species (ROS), respectively. The ELISA assay was performed to measure the caspase-2, -3, and -9 activity. ATR-FTIR spectroscopy was used to evaluate biomolecular changes in RPTECs induced by the tested formulas. The combinations of C1/NTBD and AmB did not exert a strong inhibitory effect on the viability/growth of kidney cells, as evidenced by the negligible changes in the apoptotic/necrotic rate and caspase activity, compared to the control cells. Both NTBD and C1 displayed stronger anti-oxidant activity when combined with AmB. The relatively low nephrotoxicity of the thiadiazole derivative combinations and the protective activity against AmB-induced oxidative stress may indicate their potential use in the therapy of fungal infections.
Collapse
Affiliation(s)
- Agnieszka Dróżdż
- Department of Cell Biology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Adrianna Sławińska-Brych
- Department of Cell Biology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Dominika Kubera
- Department of Cell Biology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Magdalena Kimsa-Dudek
- Department of Nutrigenomics and Bromatology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Joanna Magdalena Gola
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
- Correspondence:
| | - Jolanta Adamska
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Celina Kruszniewska-Rajs
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Arkadiusz Matwijczuk
- Department of Biophysics, University of Life Sciences, Akademicka 13, 20-950 Lublin, Poland
| | - Dariusz Karcz
- Department of Chemical Technology and Environmental Analytics, Cracow University of Technology, 31-155 Krakow, Poland
| | - Wojciech Dąbrowski
- I Clinic of Anaesthesiology and Intensive Therapy with Clinical Paediatric Department, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Mariusz Gagoś
- Department of Cell Biology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland
| |
Collapse
|
34
|
Cytotoxicity and Molecular Alterations Induced by Scorpion Venom Antimicrobial Peptide Smp43 in Breast Cancer Cell Lines MDA-MB-231 and MCF-7. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10474-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
35
|
Zhang L, Wang H, Liu J, Chen S, Yang H, Yang Z, Zhang Z, Zhao H, Yuan L, Tian L, Zhong B, Liu X. Scattering Inversion Study for Suspended Label-Free Lymphocytes with Complex Fine Structures. BME FRONTIERS 2022; 2022:9867373. [PMID: 37850176 PMCID: PMC10521707 DOI: 10.34133/2022/9867373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/28/2022] [Indexed: 10/19/2023] Open
Abstract
Objective and Impact Statement. Distinguishing malignant lymphocytes from normal ones is vital in pathological examination. We proposed an inverse light scattering (ILS) method for label-free suspended lymphocytes with complex fine structures to identify their volumes for pathological state. Introduction. Light scattering as cell's "fingerprint" provides valuable morphology information closely related to its biophysical states. However, the detail relationships between the morphology with complex fine structures and its scattering characters are not fully understood. Methods. To quantitatively inverse the volumes of membrane and nucleus as the main scatterers, clinical lymphocyte morphologies were modeled combining the Gaussian random sphere geometry algorithm by 750 reconstructed results after confocal scanning, which allowed the accurate simulation to solve ILS problem. For complex fine structures, the specificity for ILS study was firstly discussed (to our knowledge) considering the differences of not only surface roughness, posture, but also the ratio of nucleus to the cytoplasm and refractive index. Results. The volumes of membrane and nucleus were proved theoretically to have good linear relationship with the effective area and entropy of forward scattering images. Their specificity deviations were less than 3.5%. Then, our experimental results for microsphere and clinical leukocytes showed the Pearson product-moment correlation coefficients (PPMCC) of this linear relationship were up to 0.9830~0.9926. Conclusion. Our scattering inversion method could be effectively applied to identify suspended label-free lymphocytes without destructive sample pretreatments and complex experimental systems.
Collapse
Affiliation(s)
- Lu Zhang
- Xi’an Jiaotong University, School of Mechanical Engineering, State Key Laboratory for Manufacturing Systems Engineering, Xi’an 710049, China
| | - Huijun Wang
- Xi’an Jiaotong University, School of Mechanical Engineering, State Key Laboratory for Manufacturing Systems Engineering, Xi’an 710049, China
| | - Jianyi Liu
- Xi’an Jiaotong University, Institute of Artificial Intelligence and Robotics, Xi’an 710049, China
| | - Shuang Chen
- Xi’an Jiaotong University, School of Mechanical Engineering, State Key Laboratory for Manufacturing Systems Engineering, Xi’an 710049, China
| | - He Yang
- Xi’an Jiaotong University, School of Mechanical Engineering, State Key Laboratory for Manufacturing Systems Engineering, Xi’an 710049, China
| | - Zewen Yang
- Xi’an Jiaotong University, School of Mechanical Engineering, State Key Laboratory for Manufacturing Systems Engineering, Xi’an 710049, China
| | - Zhenxi Zhang
- Xi’an Jiaotong University, Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi’an 710049, China
| | - Hong Zhao
- Xi’an Jiaotong University, School of Mechanical Engineering, State Key Laboratory for Manufacturing Systems Engineering, Xi’an 710049, China
| | - Li Yuan
- Xi’an Jiaotong University, First Affiliated Hospital, Xi’an 710049, China
| | - Lifang Tian
- Xi’an Jiaotong University, Second Affiliated Hospital, Xi’an 710049, China
| | - Bo Zhong
- Xi’an Jiaotong University, Second Affiliated Hospital, Xi’an 710049, China
| | - Xiaolong Liu
- Mengchao Hepatobiliary Hospital of Fujian Medical University, The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Fuzhou 350025, China
| |
Collapse
|
36
|
Identification of Human Cell Cycle Phase Markers Based on Single-Cell RNA-Seq Data by Using Machine Learning Methods. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2516653. [PMID: 36004205 PMCID: PMC9393965 DOI: 10.1155/2022/2516653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 12/17/2022]
Abstract
The cell cycle is composed of a series of ordered, highly regulated processes through which a cell grows and duplicates its genome and eventually divides into two daughter cells. According to the complex changes in cell structure and biosynthesis, the cell cycle is divided into four phases: gap 1 (G1), DNA synthesis (S), gap 2 (G2), and mitosis (M). Determining which cell cycle phases a cell is in is critical to the research of cancer development and pharmacy for targeting cell cycle. However, current detection methods have the following problems: (1) they are complicated and time consuming to perform, and (2) they cannot detect the cell cycle on a large scale. Rapid developments in single-cell technology have made dissecting cells on a large scale possible with unprecedented resolution. In the present research, we construct efficient classifiers and identify essential gene biomarkers based on single-cell RNA sequencing data through Boruta and three feature ranking algorithms (e.g., mRMR, MCFS, and SHAP by LightGBM) by utilizing four advanced classification algorithms. Meanwhile, we mine a series of classification rules that can distinguish different cell cycle phases. Collectively, we have provided a novel method for determining the cell cycle and identified new potential cell cycle-related genes, thereby contributing to the understanding of the processes that regulate the cell cycle.
Collapse
|
37
|
Hassan RA, Hamed MI, Abdou AM, El-Dash Y. Novel antiproliferative agents bearing substituted thieno[2,3-d]pyrimidine scaffold as dual VEGFR-2 and BRAF kinases inhibitors and apoptosis inducers; design, synthesis and molecular docking. Bioorg Chem 2022; 125:105861. [DOI: 10.1016/j.bioorg.2022.105861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/05/2022] [Accepted: 05/05/2022] [Indexed: 12/15/2022]
|
38
|
Kowalczyk A, Piotrowicz M, Gapińska M, Trzybiński D, Woźniak K, Golding TM, Stringer T, Smith GS, Czerwieniec R, Kowalski K. Chemistry of glycol nucleic acid (GNA): Synthesis, photophysical characterization and insight into the biological activity of phenanthrenyl GNA constituents. Bioorg Chem 2022; 125:105847. [DOI: 10.1016/j.bioorg.2022.105847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 11/02/2022]
|
39
|
Expression and Prognostic Value of Melanoma-Associated Antigen D2 in Gliomas. Brain Sci 2022; 12:brainsci12080986. [PMID: 35892426 PMCID: PMC9330880 DOI: 10.3390/brainsci12080986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 01/10/2023] Open
Abstract
Introduction: The melanoma-associated antigen D2 (MAGED2) is one of the melanoma-associated antigen family members. It is commonly overexpressed in a variety of malignancies. However, the mechanism and function of MAGED2 in glioma remain unknown. Methods: The MAGED2 expression level and the correlations between clinical characteristics were analyzed with the data from the CGGA and TCGA datasets. MAGED2 expression in 98 glioma tissues was measured using RT-qPCR, Western blot, and immunohistochemistry. CCK-8, colony formation, and EdU assays were used to assess the effect of MAGED2 on U251-MG cell proliferation. Flow cytometry was used to track changes in the cell cycle and cell apoptosis following plasmid transfection with CRISPRi. Results: MAGED2 was shown to be highly expressed in glioma tissues, and high MAGED2 expression predicted poor prognosis. Furthermore, MAGED2 knockdown significantly inhibited the proliferation of U251-MG cells by preventing cell cycle arrest at the G0/G1 phase and triggering apoptosis. In line with in vitro findings, the results of the xenograft experiment and immunohistochemistry also showed that MAGED2 suppression inhibited tumor development and decreased Ki-67 expression levels. Conclusions: MAGED2 may be a possible biomarker for glioma and an important prognostic factor for glioma patients.
Collapse
|
40
|
Zheng CY, Chu XY, Gao CY, Hu HY, He X, Chen X, Yang K, Zhang DL. TAT&RGD Peptide-Modified Naringin-Loaded Lipid Nanoparticles Promote the Osteogenic Differentiation of Human Dental Pulp Stem Cells. Int J Nanomedicine 2022; 17:3269-3286. [PMID: 35924260 PMCID: PMC9342892 DOI: 10.2147/ijn.s371715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022] Open
Abstract
Background Naringin is a naturally occurring flavanone that promotes osteogenesis. Owing to the high lipophilicity, poor in vivo bioavailability, and extensive metabolic alteration upon administration, the clinical efficacy of naringin is understudied. Additionally, information on the molecular mechanism by which it promotes osteogenesis is limited. Methods In this study, we prepared TAT & RGD peptide-modified naringin-loaded nanoparticles (TAT-RGD-NAR-NPs), evaluated their potency on the osteogenic differentiation of human dental pulp stem cells (hDPSCs), and studied its mechanism of action through metabolomic analysis. Results The particle size and zeta potential of TAT-RGD-NAR-NPs were 160.70±2.05 mm and –20.77±0.47mV, respectively. The result of cell uptake assay showed that TAT-RGD-NAR-NPs could effectively enter hDPSCs. TAT-RGD-NAR-NPs had a more significant effect on cell proliferation and osteogenic differentiation promotion. Furthermore, in metabolomic analysis, naringin particles showed a strong influence on the glycerophospholipid metabolism pathway of hDPSCs. Specifically, it upregulated the expression of PLA2G3 and PLA2G1B (two isozymes of phospholipase A2, PLA2), increased the biosynthesis of lysophosphatidic acid (LPA). Conclusion These results suggested that TAT-RGD-NPs might be used for transporting naringin to hDPSCs for modulating stem cell osteogenic differentiation. The metabolomic analysis was used for the first time to elucidate the mechanism by which naringin promotes hDPSCs osteogenesis by upregulating PLA2G3 and PLA2G1B.
Collapse
Affiliation(s)
- Chun-Yan Zheng
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, Beijing, People’s Republic of China
| | - Xiao-Yang Chu
- Department of Stomatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Chun-Yan Gao
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, Beijing, People’s Republic of China
| | - Hua-Ying Hu
- Birth Defects Prevention and Control Technology Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Xin He
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, Beijing, People’s Republic of China
| | - Xu Chen
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, Beijing, People’s Republic of China
| | - Kai Yang
- Prenatal Diagnosis Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Dong-Liang Zhang
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, Beijing, People’s Republic of China
- Correspondence: Dong-Liang Zhang, Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, 11 Xilahutong Road, Beijing, 100040, People’s Republic of China, Email
| |
Collapse
|
41
|
Kari S, Subramanian K, Altomonte IA, Murugesan A, Yli-Harja O, Kandhavelu M. Programmed cell death detection methods: a systematic review and a categorical comparison. Apoptosis 2022; 27:482-508. [PMID: 35713779 PMCID: PMC9308588 DOI: 10.1007/s10495-022-01735-y] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2022] [Indexed: 01/15/2023]
Abstract
Programmed cell death is considered a key player in a variety of cellular processes that helps to regulate tissue growth, embryogenesis, cell turnover, immune response, and other biological processes. Among different types of cell death, apoptosis has been studied widely, especially in the field of cancer research to understand and analyse cellular mechanisms, and signaling pathways that control cell cycle arrest. Hallmarks of different types of cell death have been identified by following the patterns and events through microscopy. Identified biomarkers have also supported drug development to induce cell death in cancerous cells. There are various serological and microscopic techniques with advantages and limitations, that are available and are being utilized to detect and study the mechanism of cell death. The complexity of the mechanism and difficulties in distinguishing among different types of programmed cell death make it challenging to carry out the interventions and delay its progression. In this review, mechanisms of different forms of programmed cell death along with their conventional and unconventional methods of detection of have been critically reviewed systematically and categorized on the basis of morphological hallmarks and biomarkers to understand the principle, mechanism, application, advantages and disadvantages of each method. Furthermore, a very comprehensive comparative analysis has been drawn to highlight the most efficient and effective methods of detection of programmed cell death, helping researchers to make a reliable and prudent selection among the available methods of cell death assay. Conclusively, how programmed cell death detection methods can be improved and can provide information about distinctive stages of cell death detection have been discussed.
Collapse
Affiliation(s)
- Sana Kari
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland
| | - Kumar Subramanian
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland
| | - Ilenia Agata Altomonte
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland
| | - Akshaya Murugesan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland.,Department of Biotechnology, Lady Doak College, Thallakulam, Madurai, 625002, India
| | - Olli Yli-Harja
- Institute for Systems Biology, 1441N 34th Street, Seattle, WA, USA.,Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland. .,Department of Biotechnology, Lady Doak College, Thallakulam, Madurai, 625002, India.
| |
Collapse
|
42
|
Abdelnaby RM, El-Malah AA, FakhrEldeen RR, Saeed MM, Nadeem RI, Younis NS, Abdel-Rahman HM, El-Dydamony NM. In Vitro Anticancer Activity Screening of Novel Fused Thiophene Derivatives as VEGFR-2/AKT Dual Inhibitors and Apoptosis Inducers. Pharmaceuticals (Basel) 2022; 15:ph15060700. [PMID: 35745619 PMCID: PMC9229165 DOI: 10.3390/ph15060700] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 01/13/2023] Open
Abstract
Protein kinases are seen as promising targets in controlling cell proliferation and survival in treating cancer where fused thiophene synthon was utilized in many kinase inhibitors approved by the FDA. Accordingly, this work focused on adopting fused thienopyrrole and pyrrolothienopyrimidine scaffolds in preparing new inhibitors, which were evaluated as antiproliferative agents in the HepG2 and PC-3 cell lines. The compounds 3b (IC50 = 3.105 and 2.15 μM) and 4c (IC50 = 3.023 and 3.12 μM) were the most promising candidates on both cells with good selective toxicity-sparing normal cells. A further mechanistic evaluation revealed promising kinase inhibitory activity, where 4c inhibited VEGFR-2 and AKT at IC50 = 0.075 and 4.60 μM, respectively, while 3b showed IC50 = 0.126 and 6.96 μM, respectively. Moreover, they resulted in S phase cell cycle arrest with subsequent caspase-3-induced apoptosis. Lastly, docking studies evaluated the binding patterns of these active derivatives and demonstrated a similar fitting pattern to the reference ligands inside the active sites of both VEGFR-2 and AKT (allosteric pocket) crystal structures. To conclude, these thiophene derivatives represent promising antiproliferative leads inhibiting both VEGFR-2 and AKT and inducing apoptosis in liver cell carcinoma.
Collapse
Affiliation(s)
- Rana M. Abdelnaby
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
- Correspondence: (R.M.A.); (N.M.E.-D.); Tel.: +20-01001797688 or +2-01270551779 (R.M.A.)
| | - Afaf A. El-Malah
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Rasha R. FakhrEldeen
- Biochemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12585, Egypt;
| | - Marwa M. Saeed
- Pharmacology and Toxicology Lecturer, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt;
| | - Rania I. Nadeem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt;
| | - Nancy S. Younis
- Pharmaceutical Sciences Department, Faculty of Clinical Pharmacy, King Faisal University, Al-Ahsa, Al-Hofuf 31982, Saudi Arabia;
| | - Hanaa M. Abdel-Rahman
- Pharmacy Practice Department, Faculty of Pharmacy, Egyptian Russian University, Cairo 11829, Egypt;
- Department of Forensic Medicine and Toxicology, Faculty of Medicine, Ain Shams University, Cairo 11562, Egypt
| | - Nehad M. El-Dydamony
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12585, Egypt
- Correspondence: (R.M.A.); (N.M.E.-D.); Tel.: +20-01001797688 or +2-01270551779 (R.M.A.)
| |
Collapse
|
43
|
TRAIL/S-layer/graphene quantum dot nanohybrid enhanced stability and anticancer activity of TRAIL on colon cancer cells. Sci Rep 2022; 12:5851. [PMID: 35393438 PMCID: PMC8991220 DOI: 10.1038/s41598-022-09660-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL), known as a cytokine of the TNF superfamily, is considered a promising antitumor agent due to its ability to selectively induce apoptosis in a wide variety of cancer cells. However, failure of its successful translation into clinic has led to development of nano-based platforms aiming to improve TRAIL therapeutic efficacy. In this regard, we fabricated a novel TRAIL-S-layer fusion protein (S-TRAIL) conjugated with graphene quantum dots (GQDs) to benefit both the self-assembly of S-layer proteins, which leads to elevated TRAIL functional stability, and unique optical properties of GQDs. Noncovalent conjugation of biocompatible GQDs and soluble fusion protein was verified via UV–visible and fluorescence spectroscopy, size and ζ-potential measurements and transmission electron microscopy. The potential anticancer efficacy of the nanohybrid system on intrinsically resistant cells to TRAIL (HT-29 human colon carcinoma cells) was investigated by MTT assay and flow cytometry, which indicated about 80% apoptosis in cancer cells. These results highlight the potential of TRAIL as a therapeutic protein that can be extensively improved by taking advantage of nanotechnology and introduce S-TRAIL/GQD complex as a promising nanohybrid system in cancer treatment.
Collapse
|
44
|
Abdelnaby RM, Rateb HS, Ali O, Saad AS, Nadeem RI, Abou-Seri SM, Amin KM, Younis NS, Abdelhady R. Dual PI3K/Akt Inhibitors Bearing Coumarin-Thiazolidine Pharmacophores as Potential Apoptosis Inducers in MCF-7 Cells. Pharmaceuticals (Basel) 2022; 15:ph15040428. [PMID: 35455425 PMCID: PMC9027131 DOI: 10.3390/ph15040428] [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: 01/18/2022] [Revised: 02/05/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Breast cancer is the most common malignancy worldwide; therefore, the development of new anticancer agents is essential for improved tumor control. By adopting the pharmacophore hybridization approach, two series of 7-hydroxyl-4-methylcoumarin hybridized with thiosemicarbazone (V–VI) and thiazolidin-4-one moieties (VII–VIII) were prepared. The in vitro anticancer activity was assessed against MCF-7 cells adopting the MTT assay. Nine compounds showed significant cytotoxicity. The most promising compound, VIIb, induced remarkable cytotoxicity (IC50 of 1.03 + 0.05 µM). Further investigations were conducted to explore its pro-apoptotic activity demonstrating S-phase cell cycle arrest. Apoptosis rates following VIIb treatment revealed a 5-fold and 100-fold increase in early and late apoptotic cells, correspondingly. Moreover, our results showed caspase-9 dependent apoptosis induction as manifested by an 8-fold increase in caspase-9 level following VIIb treatment. Mechanistically, VIIb was found to target the PI3K-α/Akt-1 axis, as evidenced by enzyme inhibition assay results reporting significant inhibition of examined enzymes. These findings were confirmed by Western blot results indicating the ability of VIIb to repress levels of Cyclin D1, p-PI3K, and p-Akt. Furthermore, docking studies showed that VIIb has a binding affinity with the PI3K binding site higher than the original ligands X6K. Our results suggest that VIIb has pharmacological potential as a promising anti-cancer compound by the inhibition of the PI3K/Akt axis.
Collapse
Affiliation(s)
- Rana M. Abdelnaby
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
- Correspondence: ; Tel.: +20-1270551779
| | - Heba S. Rateb
- Pharmaceutical Chemistry Department, Faculty of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12585, Egypt;
| | - Omaima Ali
- Egyptian Drug Authority, Cairo 12618, Egypt;
| | - Ahmed S. Saad
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Port Said University, Port Said 42511, Egypt;
| | - Rania I. Nadeem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt;
| | - Sahar M. Abou-Seri
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; (S.M.A.-S.); (K.M.A.)
| | - Kamilia M. Amin
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; (S.M.A.-S.); (K.M.A.)
| | - Nancy S. Younis
- Pharmaceutical Sciences Department, Faculty of Clinical Pharmacy, King Faisal University, Al Hofuf 31982, Al-Ahsa, Saudi Arabia;
| | - Rasha Abdelhady
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum 63514, Egypt;
| |
Collapse
|
45
|
El-Dydamony NM, Abdelnaby RM, Abdelhady R, Ali O, Fahmy MI, R. Fakhr Eldeen R, Helwa AA. Pyrimidine-5-carbonitrile based potential anticancer agents as apoptosis inducers through PI3K/AKT axis inhibition in leukaemia K562. J Enzyme Inhib Med Chem 2022; 37:895-911. [PMID: 35345960 PMCID: PMC8967206 DOI: 10.1080/14756366.2022.2051022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A novel series of 4-(4-Methoxyphenyl)-2-(methylthio)pyrimidine-5-carbonitrile was developed linked to an aromatic moiety via N-containing bridge and then evaluated for their cytotoxic activity against MCF-7 and K562 cell lines. Seven compounds exhibited the highest activity against both cell lines where compounds 4d and 7f were the most active against K562 cell line. Exploring their molecular mechanisms by enzyme inhibition assay on PI3Kδ/γ and AKT-1 showed that compound 7f was promising more than 4d with IC50 = 6.99 ± 0.36, 4.01 ± 0.55, and 3.36 ± 0.17 uM, respectively. Also, flowcytometric analysis revealed that 7f caused cell cycle arrest at S-phase followed by caspase 3 dependent apoptosis induction. Mechanistically, compound 7f proved to modulate the expression of PI3K, p-PI3K, AKT, p-AKT, Cyclin D1, and NFΚβ. Furthermore, in-vivo toxicity study indicated good safety profile for 7f. These findings suggest that the trimethoxy derivative 7f has strong potential as a multi-acting inhibitor on PI3K/AKT axis targeting breast cancer and leukaemia.
Collapse
Affiliation(s)
- Nehad M. El-Dydamony
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Rana M. Abdelnaby
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Rasha Abdelhady
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Omaima Ali
- Cell Line Unit, Egyptian Drug Authority (EDA), Cairo, Egypt
| | - Mohamed I. Fahmy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Rasha R. Fakhr Eldeen
- Biochemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Amira A. Helwa
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| |
Collapse
|
46
|
Duarte SS, Silva DKF, Lisboa TMH, Gouveia RG, de Andrade CCN, de Sousa VM, Ferreira RC, de Moura RO, Gomes JNS, da Silva PM, de Lourdes Assunção Araújo de Azevedo F, Keesen TSL, Gonçalves JCR, Batista LM, Sobral MV. Apoptotic and antioxidant effects in HCT-116 colorectal carcinoma cells by a spiro-acridine compound, AMTAC-06. Pharmacol Rep 2022; 74:545-554. [PMID: 35297003 DOI: 10.1007/s43440-022-00357-0] [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: 10/27/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Acridine compounds have been described as promising anticancer agents. Previous studies showed that (E)-1'-((4-chlorobenzylidene)amino)-5'-oxo-1',5'-dihydro-10H-spiro[acridine-9,2'-pyrrole]-4'-carbonitrile (AMTAC-06), a spiro-acridine compound, has antitumor activity on Ehrlich tumor and low toxicity. Herein, we investigated its antitumor effect against human cells in vitro. METHODS MTT assay was used to assess cytotoxicity of AMTAC-06 (3.125-200 µM) against tumor and non-tumor cells, and the half-maximal inhibitory concentration (IC50) and the selectivity index (SI) were calculated. The effects on the cell cycle (propidium iodide-PI-staining), apoptosis (Annexin V-FITC/PI double staining by flow cytometry), and production of reactive oxygen species, ROS (DCFH assay) were also evaluated. Statistical analysis was achieved using ANOVA followed by Tukey's post-test. RESULTS AMTAC-06 showed higher cytotoxicity against colorectal carcinoma HCT-116 cells (IC50: 12.62 µM). The SI showed that AMTAC-06 was more selective for HCT-116 cells (HaCaT SI: 1.41; PBMC SI: 0.62) than doxorubicin (HaCaT SI: 0.10; PBMC SI: 0.01). AMTAC-06 (15 and 30 µM) induced an increase in the sub-G1 peak (p < 0.000001) and cell cycle arrest in S phase (p = 0.003547). Moreover, treatment with this compound (15 and 30 µM) resulted in increased early (p < 0.000001) and late apoptotic cells (p < 0.000001). In addition, there was a reduction on ROS production (p < 0.000001). CONCLUSIONS AMTAC-06 presents anticancer activity against HCT-116 cells by regulating the cell cycle, inducing apoptosis and an antioxidant action.
Collapse
Affiliation(s)
- Sâmia Sousa Duarte
- Postgraduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Daiana Karla Frade Silva
- Postgraduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Thaís Mangeon Honorato Lisboa
- Postgraduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Rawny Galdino Gouveia
- Postgraduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | | | - Valgrícia Matias de Sousa
- Postgraduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Rafael Carlos Ferreira
- Postgraduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Ricardo Olimpio de Moura
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Joilly Nilce Santana Gomes
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Patricia Mirella da Silva
- Invertebrate Immunology and Pathology Laboratory, Department of Molecular Biology, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | | | - Tatjana S L Keesen
- Immunology of Infectious Diseases Laboratory, Biotechnology Center, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | | | - Leônia Maria Batista
- Postgraduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Paraíba, Brazil.,Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Marianna Vieira Sobral
- Postgraduation Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa, Paraíba, Brazil. .,Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Paraíba, Brazil. .,Laboratório de Oncofarmacologia (Oncofar), Instituto de Pesquisa em Fármacos e Medicamentos (IPeFarM). Cidade Universitária, Campus I, João Pessoa, Paraíba, 58051-900, Brazil.
| |
Collapse
|
47
|
Wang X, Tian X, Sui X, Li X, Zhao X, Han K, Sun L, Dong Y. Increased expression of NCAPG (Non-SMC condensing I complex subunit G) is associated with progression and poor prognosis of lung adenocarcinoma. Bioengineered 2022; 13:6113-6125. [PMID: 35254214 PMCID: PMC8974211 DOI: 10.1080/21655979.2022.2035124] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/09/2021] [Accepted: 12/08/2021] [Indexed: 01/04/2023] Open
Abstract
Recently, studies have shown that the up-regulation of Non-SMC Condensin I Complex Subunit G (NCAPG) in some tumors can promote tumor progression, and its high expression has a strong correlation with the poor prognosis of patients. However, there are few studies on NCAPG in lung adenocarcinoma (LUAD). Our research is to explore the role of NCAPG in LUAD and try to reveal the possible molecular mechanism. We use public databases and tissue samples from LUAD patients to verify that NCAPG is significantly up-regulated in LUAD, and the high expression of NCAPG is related to the poor prognosis of patients. Subsequently, we found that silencing NCAPG can inhibit the proliferation and invasion of LUAD cells in vitro and the growth of subcutaneous tumors in nude mice in vivo. In order to explore the possible molecular mechanism of NCAPG's function, we found out the genes co-expressed with NCAPG through the cBioportal database, and discovered that these genes were significantly enriched in the cell cycle and other pathways through DAVID analysis, which implies the importance of NCAPG in the cell cycle. Finally, we confirmed by flow cytometry that NCAPG affects the conversion of cell cycle mitosis from G1 to S. Taken together, our research results suggest that NCAPG plays a role in the progress of LUAD. Moreover, NCAPG can be used as a potential biomarker for the diagnosis of LUAD, as well as a potential therapeutic target for patients with LUAD.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of Oncology, Zibo City Fourth People’s Hospital, Zibo, Shandong, China
| | - Xia Tian
- Department of Oncology, Zibo City Fourth People’s Hospital, Zibo, Shandong, China
| | - Xufang Sui
- Department of Breast and Thyroid Surgery, Zibo Central Hospital, Zibo, China
| | - Xiangfeng Li
- Department of Radiology, Zibo City Fourth People’s Hospital, Zibo, Shandong, China
| | - Xiaoyang Zhao
- Department of Surgery, Zibo City Fourth People’s Hospital, Zibo, Shandong, China
| | - Kai Han
- Department of Internal Medicine, Zibo City Fourth People’s Hospital, Zibo, Shandong, China
| | - Lingyan Sun
- Department of Oncology, Zibo Central Hospital, Zibo, China
| | - Yujin Dong
- Department of Oncology, Zibo Central Hospital, Zibo, China
| |
Collapse
|
48
|
Punpai S, Saenkham A, Jarintanan F, Jongrungruangchok S, Choowongkomon K, Suksamrarn S, Tanechpongtamb W. HDAC inhibitor cowanin extracted from G. fusca induces apoptosis and autophagy via inhibition of the PI3K/Akt/mTOR pathways in Jurkat cells. Biomed Pharmacother 2022; 147:112577. [PMID: 35078092 DOI: 10.1016/j.biopha.2021.112577] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
Cowanin, a xanthone derivative extracted from the Garcinia fusca plant, has been recognized for various biological activities including, antimicrobial, anti-inflammatory, and anticancer activities. However, the mechanism to induce cancer cell death in cancer cells remains to be fully elucidated. Our previous report showed that other xanthones from these plants could act as histone deacetylase inhibitors (HDACi), so we deeply analyzed the role of cowanin, a major compound of G.fusca, and investigated through the mode of cell death both apoptosis and autophagy that have never been reported. As a result, it was demonstrated that cowanin indicated the role of HDACi as other xanthones. The molecular docking analysis showed that cowanin could interact within the catalytic pocket region of HDAC class I (HDAC2, 8) and II (HDAC4, 7) proteins and inhibit their activity. Also, the level of protein expression of HDAC2, 4, 7, and 8 was distinctly decreased, and the level of histone H3 and H4 acetylation increased in cowanin treated cells. For the mode of cell death, cowanin demonstrated both apoptosis and autophagy activation in Jurkat cells. Besides, cowanin significantly suppressed phosphorylation of PI3K, Akt, and mTOR signaling. Therefore, these findings revealed that cowanin represents a new promising candidate for development as an anticancer agent by inducing apoptosis and autophagy via PI3K/AKT/mTOR pathway and effectively inhibiting HDAC activity.
Collapse
Affiliation(s)
- Sakdiphong Punpai
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Audchara Saenkham
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Srinakharinwirot University, Bangkok 10110, Thailand
| | | | | | - Kiattawee Choowongkomon
- Departmentof Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10903, Thailand
| | - Sunit Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Wanlaya Tanechpongtamb
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand.
| |
Collapse
|
49
|
Yuan HH, Zhang XC, Wei XL, Zhang WJ, Du XX, Huang P, Chen H, Bai L, Zhang HF, Han Y. LncRNA UCA1 mediates Cetuximab resistance in Colorectal Cancer via the MiR-495 and HGF/c-MET Pathways. J Cancer 2022; 13:253-267. [PMID: 34976187 PMCID: PMC8692674 DOI: 10.7150/jca.65687] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Cetuximab is one of the most widely used monoclonal antibodies to treat patients with RAS/BRAF wild-type metastatic colorectal cancer (mCRC). Unfortunately, cetuximab resistance often occurs during targeted therapy. However, the underlying epigenetic mechanisms remain unclear. Our previous study demonstrated that the exosomal transfer of urothelial carcinoma-associated 1 (UCA1) confers cetuximab resistance to CRC cells. The goal of this study was to elucidate the detailed role of UCA1 in cetuximab resistance in CRC and the underlying molecular mechanism. Methods:In vitro and in vivo functional studies were performed to assess the role of UCA1 in cetuximab resistance in CRC cell lines and xenograft models. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was used to examine UCA1 localization and expression. Bioinformatics analysis was performed to predict the potential mechanism of UCA1, which was further validated by the dual-luciferase reporter assay and the RNA immunoprecipitation (RIP) assay. Cells treated with indicators were subjected to Cell Counting Kit-8 (CCK-8) and western blotting to investigate the role of hepatocyte growth factor (HGF)/c-mesenchymal-epithelial transition (c-MET) signalling in UCA1-mediated cetuximab resistance. Results: We showed that UCA1 decreased CRC cell sensitivity to cetuximab by suppressing apoptosis. Mechanistic studies revealed that UCA1 promoted cetuximab resistance by competitively binding miR-495 to facilitate HGF and c-MET expression in CRC cells. Moreover, HGF was shown to attenuate the cetuximab-induced inhibition of cell proliferation by activating the HGF/c-MET pathway in CRC cells. Conclusion: We provide the first evidence of a UCA1-miR-495-HGF/c-MET regulatory network involved in cetuximab resistance in CRC. Therefore, UCA1 has potential as a predictor and therapeutic target for cetuximab resistance.
Collapse
Affiliation(s)
- Heng-Heng Yuan
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Xin-Chen Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiao-Li Wei
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Wen-Jie Zhang
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Xiao-Xue Du
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Peng Huang
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Hao Chen
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Lu Bai
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Hong-Feng Zhang
- Department of Gastric Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Yu Han
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| |
Collapse
|
50
|
Abdel Ghany LMA, El-Dydamony NM, Helwa AA, Abdelraouf SM, Abdelnaby RM. Coumarin-acetohydrazide derivatives as novel antiproliferative agents via VEGFR-2/AKT axis inhibition and apoptosis triggering. NEW J CHEM 2022. [DOI: 10.1039/d2nj02436e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The VEGFR-2/AKT pathway is a crucial axis in tumor survival where it is highly dysregulated in many cancer types.
Collapse
Affiliation(s)
- Lina M. A. Abdel Ghany
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Nehad M. El-Dydamony
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Amira A. Helwa
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Sahar M. Abdelraouf
- Biochemistry Department, Faculty of pharmacy, Misr International University, Cairo, Egypt
| | - Rana M. Abdelnaby
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|