1
|
Park SB, Choi HY, Park YE, Jang S, Chun HS. High-content screening morphological analysis to evaluate hepatic apoptosis induced by plant alkaloids in a Chang cell model. Toxicology 2025; 515:154140. [PMID: 40222580 DOI: 10.1016/j.tox.2025.154140] [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: 02/07/2025] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
As interest in plant-derived compounds and their application in the pharmaceutical and functional food industries has increased, the rapid detection of chemical toxicity has become increasingly important for developing safe products. High-content screening (HCS) can quantify cellular and organelle morphological changes through image analysis; however, most HCS studies on apoptosis, a key toxicological event, have focused on the expression of apoptosis-related genes or proteins. In this study, we aimed to verify whether apoptosis can be detected solely based on cellular morphological changes. Chang cells were treated with staurosporine (STS), a well-known apoptosis inducer, and the morphological changes in the cells were quantified using HCS assays. The correlation between these HCS morphological descriptors and apoptosis rates measured using flow cytometry was determined. Chang cells were also treated with several plant-derived alkaloids known to induce apoptosis, and the same process was performed. The correlation coefficients, which were used to evaluate the correlation between HCS descriptors and apoptosis rates after STS treatment, ranged from 0.64 to 0.98, with 13 descriptors showing significant correlations. In contrast, the highest correlation coefficients between HCS descriptors and apoptosis rates after treatment with 1 of the 12 alkaloids investigated were determined to be 0.75 (at 10 μg/ml) and 0.49 (at 100 μg/ml). The apoptosis-related morphological changes induced by STS and alkaloids were observed using confocal microscopy. The present study demonstrates that HCS assays can detect apoptosis solely based on cellular morphological changes, providing a potential tool for rapid toxicity screening in early product development.
Collapse
Affiliation(s)
- Su Been Park
- GreenTech-based Food Safety Research Group, BK21 Four, School of Food Science and Technology, Chung-Ang University, South Korea.
| | - Hwa Young Choi
- GreenTech-based Food Safety Research Group, BK21 Four, School of Food Science and Technology, Chung-Ang University, South Korea
| | - Young Eun Park
- GreenTech-based Food Safety Research Group, BK21 Four, School of Food Science and Technology, Chung-Ang University, South Korea
| | - Sihyeon Jang
- GreenTech-based Food Safety Research Group, BK21 Four, School of Food Science and Technology, Chung-Ang University, South Korea.
| | - Hyang Sook Chun
- GreenTech-based Food Safety Research Group, BK21 Four, School of Food Science and Technology, Chung-Ang University, South Korea.
| |
Collapse
|
2
|
Ashraf R, Khalid Z, Qin QP, Iqbal MA, Taskin-Tok T, Bayil İ, Quah CK, Daud NAM, Alqahtany FZ, Amin MA, El-Bahy SM. Synthesis of N-heterocyclic carbene‑selenium complexes modulating apoptosis and autophagy in cancer cells: Probing the interactions with biomolecules and enzymes. Bioorg Chem 2025; 160:108435. [PMID: 40199010 DOI: 10.1016/j.bioorg.2025.108435] [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: 02/14/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
Growing cancer resistance is a global threat that calls for development of newer chemotherapeutic analogues especially targeted based therapy to enhance efficacy and selectivity. In this contribution, herein, we report synthesis of selenium incorporated N-heterocyclic carbene (NHC) compounds to explore their potential cytotoxicity against HeLa cells. Test compounds were assured for suitability as drug candidates through physiochemical properties that showed lipophilicity logP 0.85-1.45 for C1-C3 and found stable in biological media (DMEM), whereas, least reactive with N-acetyl cysteine (NAC) and L-glutathione. All the studied compounds showed good cytotoxicity against various cancer strains while compound C1 [3,3-(hexane-1,6-diyl)bis(1-phenethyl-1H-imidazole-2(3H)-selenone)] and C2 [3,3-(hexane-1,6-diyl)bis(1-decyl-1H-imidazole-2(3H)-selenone)] showed promising results with IC50 values of 14.65 ± 0.66 and 8.05 ± 0.35 μg/mL respectively as compared to positive control 21.5 ± 0.05 μg/mL against HeLa cell lines. These compounds showed six-fold higher apoptosis than control with higher accumulation of Ca+ ions intracellularly that alters the expression level of autophagy proteins and increased capase-9 activity. Cell cycle analysis indicated an arrest of cycle in G1 phase of HeLa cells when treated with C1 & C2. Test compounds showed prominent affinity for binding with DNA and inhibiting thioredoxin reductase enzymes in time dependent manners. These findings indicate that Selenium NHC compounds are promising drug candidates to induce cytotoxicity via apoptosis, autophagy and mitochondrial membrane disruptions to manage tumor growth.
Collapse
Affiliation(s)
- Rizwan Ashraf
- Department of Chemistry, University of Agriculture Faisalabad, Pakistan.
| | - Zohra Khalid
- Department of Chemistry, University of Agriculture Faisalabad, Pakistan
| | - Qi-Pin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, School of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China
| | - Muhammad Adnan Iqbal
- Department of Chemistry, University of Agriculture Faisalabad, Pakistan; Organometallic and Coordination Chemistry Laboratory, Department of Chemistry, University of Agriculture Faisalabad, 38000, Pakistan.
| | - Tugba Taskin-Tok
- Faculty of Arts and Sciences, Department of Chemistry, Gaziantep University, +9027310, Gaziantep, Turkiye; Institute of Health Sciences, Department of Bioinformatics and Computational Biology, Gaziantep University, +9027310, Gaziantep, Turkiye
| | - İmren Bayil
- Institute of Health Sciences, Department of Bioinformatics and Computational Biology, Gaziantep University, +9027310, Gaziantep, Turkiye
| | - Ching Kheng Quah
- X-ray Crystallography Unit, School of Physics, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Nur Aisyah Mohamad Daud
- X-ray Crystallography Unit, School of Physics, Universiti Sains Malaysia, Penang 11800, Malaysia
| | | | - Mohammed A Amin
- Department of chemistry, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Salah M El-Bahy
- Department of chemistry, Turabah University college, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
3
|
Ryan P, Lee J. In vitro senescence and senolytic functional assays. Biomater Sci 2025. [PMID: 40375674 DOI: 10.1039/d4bm01684j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
A detailed understanding of aging biology and the development of anti-aging therapeutic strategies remain imperative yet inherently challenging due to the protracted nature of aging. Cellular senescence arises naturally through replicative exhaustion and is accelerated by clinical treatments or environmental stressors. The accumulation of senescent cells-defined by a loss of mitogenic potential, resistance to apoptosis, and acquisition of a pro-inflammatory secretory phenotype-has been implicated as a key driver of chronic disease, tissue degeneration, and organismal aging. Recent studies have highlighted the therapeutic promise of senolytic drugs, which selectively eliminate senescent cells. Compelling results from preclinical animal studies and ongoing clinical trials underscore this potential. However, the clinical translation of senolytics requires further pharmacological validation to refine selectivity, minimize toxicity, and determine optimal dosing. Equally important is the evaluation of senolytics' potential to restore tissue structure and function by reducing the senescent cell burden. In vitro tissue culture models offer a powerful platform to advance these efforts. This review summarizes the current landscape of in vitro systems used for inducing cellular senescence-referred to as "senescence assays"-and for screening senolytic drugs-referred to as "senolytic assays". We conclude by discussing key challenges to improving mechanistic insight, predictive accuracy, and clinical relevance in senolytic drug development, as well as emerging applications of senolytic therapies.
Collapse
Affiliation(s)
- Patrick Ryan
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, 01003, USA.
| | - Jungwoo Lee
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, 01003, USA.
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
4
|
Venkatesan KB, Alamelu S, Srinivasan MK, Pachaiappan P. Nerolidol loaded beta cyclodextrin nanoparticles: a promising strategy for inducing apoptosis in breast cancer cells (MCF-7). JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-31. [PMID: 40326384 DOI: 10.1080/09205063.2025.2491605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/02/2025] [Indexed: 05/07/2025]
Abstract
This study investigates the synthesis, characterization and anticancer efficacy of nerolidol-loaded beta cyclodextrin polymeric nanoparticles (NER-βCD-NPs) against MCF-7 breast cancer cells. Nerolidol, a sesquiterpene with anti-inflammatory, antioxidant, antimicrobial and anticancer properties, faces challenges of poor solubility and bioavailability, limiting its therapeutic potential. Breast cancer, a leading cause of cancer-related deaths in women, necessitates alternative therapies with fewer side effects compared to conventional chemotherapy. NER-βCD-NPs were synthesized and characterized using UV-visible spectroscopy, fourier transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), dynamic light scattering (DLS), zeta potential analysis and differential scanning calorimetry (DSC). Drug encapsulation efficiency and in vitro release were analyzed using HPLC, while molecular docking assessed NER-βCD interactions. Characterization confirmed successful nanoparticle synthesis. UV-visible spectra and FTIR indicated encapsulation-specific changes, SEM revealed surface morphology, and DLS, zeta potential and DSC analyses demonstrated increased size and stability. The encapsulation efficiency was 84.9%, with 86% NER release at pH 5.4 over 48 h. Docking studies supported strong binding between NER and βCD (binding energy: -3.55 kcal/mol). Cytotoxicity assays showed significant MCF-7 cell inhibition. Mechanistic studies revealed reactive oxygen species (ROS) generation, mitochondrial dysfunction, nuclear changes and cell cycle arrest in the G0-G1 phase. Molecular analysis demonstrated apoptosis through upregulation of Bax, Caspase 6, Caspase 9 and Cytochrome c, alongside Bcl-2 downregulation. These results highlight NER-βCD-NPs as a promising strategy for breast cancer therapy, offering targeted delivery and enhanced therapeutic efficacy while mitigating nerolidol limitations. Further studies are warranted to validate their potential in clinical applications.
Collapse
Affiliation(s)
- Kamalesh Balakumar Venkatesan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Saravanan Alamelu
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Manoj Kumar Srinivasan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Pugalendhi Pachaiappan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| |
Collapse
|
5
|
Purohit SS, Biswal A, Mohapatra P, Khamari O, Dash K, Mishra M, Biswal SB, Nayak S, Swain SK. Lysozyme/N-GQD loaded carboxymethyl cellulose hydrogels for healing of excision wounds in Drosophila and Sprague Dawley rats. Int J Biol Macromol 2025; 306:141638. [PMID: 40037441 DOI: 10.1016/j.ijbiomac.2025.141638] [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: 12/16/2024] [Revised: 02/17/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Delayed healing and fibrosis at the wound site present significant challenges in the wound care industry, often leading to complications such as infections, chronic wounds, and impaired tissue regeneration. Therefore, there is a critical need for advanced wound dressing materials that promote faster healing, prevent bacterial infections, and support effective tissue repair. This study aims to develop a Lysozyme (Lys)-based wound dressing with enhanced wound closure rates by incorporating nitrogen-doped graphene quantum dots (N-GQDs) as a functionalized nanofiller to strengthen its antibacterial properties. The wound dressing, formulated with a carboxymethyl cellulose (CMC) crosslinked polyvinylpyrrolidone (PVP) matrix, creates a porous structure that enhances swelling capacity and water vapor transmission rates (WVTR), while cytotoxicity studies confirm its biocompatibility, showing 100 % cell viability in HCT 116 and MCF7 cell lines. The in vivo wound healing performance of the designed nanocomposite hydrogel reflects complete wound closure in 5 h for Drosophila Melanogaster, aided by the shorter life span and faster metabolic processes in Drosophila, and 14 days in Sprague Dawley rat models. These results qualify the material as a promising candidate for wound dressing applications, bridging the gap between material science and medical science for effective wound management.
Collapse
Affiliation(s)
- Shuvendu Shuvankar Purohit
- Department of Chemistry, Veer Surendra Sai University of Technology, Burla, Sambalpur 768018, Odisha, India
| | - Anuradha Biswal
- Department of Chemistry, Veer Surendra Sai University of Technology, Burla, Sambalpur 768018, Odisha, India
| | - Priyaranjan Mohapatra
- Department of Chemistry, Veer Surendra Sai University of Technology, Burla, Sambalpur 768018, Odisha, India
| | - Ojaswini Khamari
- Department of Chemistry, Veer Surendra Sai University of Technology, Burla, Sambalpur 768018, Odisha, India
| | - Kalpanarani Dash
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Monalisa Mishra
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Sashi Bhusan Biswal
- Department of Pharmacology, Veer Surendra Sai Institute of Medical Science and Research, Burla, Sambalpur 768018, Odisha, India
| | - Sunanda Nayak
- Department of Phathology, Veer Surendra Sai Institute of Medical Science and Research, Burla, Sambalpur 768018, India
| | - Sarat K Swain
- Department of Chemistry, Veer Surendra Sai University of Technology, Burla, Sambalpur 768018, Odisha, India.
| |
Collapse
|
6
|
Ensoy M, Parıltı DN, Alkan AH, İlhan KNK, Mutlu P, Dedeoğlu BG, Cansaran‐Duman D. Evernic Acid: A Low-Toxic and Selective Alternative to Chemotherapeutic Agents in the Treatment of Ovarian Cancer. Arch Pharm (Weinheim) 2025; 358:e70015. [PMID: 40405479 PMCID: PMC12099196 DOI: 10.1002/ardp.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025]
Abstract
Evernic acid (EA) has emerged as a potential therapeutic agent with its low toxicity and anticancer properties. In this study, the anticancer effect of EA on ovarian cancer cell lines and normal ovarian surface epithelial cells (OSE) was evaluated. The antiproliferative effect of EA was evaluated by xCELLigence Real-Time Cell analysis, colony formation assay, and acridine orange and DAPI staining methods. Genotoxicity analysis was performed by comet assay. The effect of EA on cell migration was analyzed by wound healing assay. The potential of EA to induce apoptosis was also determined by evaluating the changes in gene and protein expression levels by qRT-PCR and Western blot analysis, respectively. EA was found to be a promising potential therapeutic agent for ovarian cancer without showing significant cytotoxic effect on normal cells. Furthermore, EA decreased the ability of ovarian cancer cells for migration, increased the rate of apoptosis by inhibiting BIRC5 and activating CASP3, triggered cell cycle arrest in the G2/M phase, and caused a decrease in mitochondrial membrane potential and genotoxic effects. The results have shown that EA could be an effective candidate molecule for ovarian cancer treatment.
Collapse
Affiliation(s)
- Mine Ensoy
- Biotechnology InstituteAnkara UniversityKeçiörenAnkaraTürkiye
| | | | - Ayşe Hale Alkan
- Biotechnology InstituteAnkara UniversityKeçiörenAnkaraTürkiye
| | | | - Pelin Mutlu
- Biotechnology InstituteAnkara UniversityKeçiörenAnkaraTürkiye
| | | | | |
Collapse
|
7
|
Kalındemirtaş FD, Cilasun GE, Kariper A. Enhanced therapeutic efficacy of platinum-doxorubicin nanoparticles on colon and breast cancer cell lines. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04080-4. [PMID: 40299021 DOI: 10.1007/s00210-025-04080-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025]
Abstract
In this study, platinum nanoparticles (PtNPs) were synthesized and their potential to improve the efficacy of doxorubicin (DOX) in cancer treatment was investigated. H2PtCl6, LiAlH4, and trisodium citrate were used during the synthesis of PtNPs. They were characterized using dynamic light scattering (DLS), Fourier transform infrared spectroscopy (FTIR), and scanning transmission electron microscopy (STEM). The diameter of the PtNPs was measured to be 21.72 nm without DOX loading and approximately 212 nm after DOX loading (DOX-PtNPs). FTIR confirmed the binding of DOX to PtNPs. In addition, MTT assays showed that DOX-PtNPs have a stronger effect on MCF-7 and HCT116 cancer cells than free DOX, even at low doses. The IC50 value for MCF-7 cells treated with DOX was determined to be 4.81 µg/ml, while it was significantly lower for the DOX-PtNP group at 0.64 µg/ml. A similar trend was observed in HCT116 cells, where the IC50 value for DOX was 5.03 µg/ml, while for DOX-PtNPs it was 0.62 µg/ml. In summary, the activity of DOX in these cells was increased approximately eightfold by PtNPs. Moreover, DOX-PtNPs showed no significant cytotoxic effects on normal HUVEC cells at low doses. Moreover, DOX-PtNPs enhanced apoptotic activity in HCT116 cells without inducing drug resistance as demonstrated by Rho123 staining and annexin/PI analyses. The significance of this study lies in the pioneering use of DOX-PtNPs in colon cancer, the synthesis of smaller PtNPs, the eightfold increase in the efficacy of DOX, and the demonstration that DOX-PtNPs do not significantly increase drug resistance.
Collapse
Affiliation(s)
| | - Gökçe Erdemir Cilasun
- Department of Medical Biology, Faculty of Medicine, Biruni University, Istanbul, Turkey
| | - Afşin Kariper
- Department of Science Education, Education Faculty, Erciyes University, Kayseri, Turkey
| |
Collapse
|
8
|
Zhang X, Sathiyaseelan A, Jin T, Wang MH. Synthesis of folic acid-tailored chitosan-coated exosomes for targeted delivery of 5-fluorouracil to triple-negative breast cancer cells. Colloids Surf B Biointerfaces 2025; 253:114737. [PMID: 40328147 DOI: 10.1016/j.colsurfb.2025.114737] [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: 10/29/2024] [Revised: 03/27/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
The use of anticancer drugs is integral to cancer treatment programs. However, the drawbacks of these chemotherapeutic agents, coupled with the problem of drug resistance, remain significant challenges. To address this, we developed a drug delivery platform based on exosomes derived from HEK293 cells, combined with folic acid-conjugated chitosan (FA-CS). The formulation, FA-CS-PEG-5FU@HEK-EXs, exhibited a polydispersity index (PDI) of 0.140, a zeta size of 188.30 nm, and a zeta potential of 3.60 mV. Its cytotoxicity to healthy tissue was negligible; however, at a dose of 500 μg/mL, the survival rate of breast cancer MDA-MB-231 cells decreased to approximately 50 %. Fluorescence staining indicated that FA-CS-PEG-5FU@HEK-EXs induced cell death in cancer cells by increasing reactive oxygen species levels, compromising the mitochondrial membrane potential, and nucleus. Furthermore, FA-CS demonstrated synergistic effects with 5FU, inducing the necrotic cell death (44.6 %). In conclusion, this study demonstrates that using exosomes to deliver the anticancer drug 5FU enhances the drug's therapeutic efficacy. Moreover, compared to conventional cancer therapies, FA-CS-PEG-5FU@HEK-EXs can minimize systemic side effects in clinical applications while enhancing drug utilization, stability, and cellular uptake, leading to highly effective treatment outcomes. The safe and efficient exosome-based platform with significant potential to inhibit tumor proliferation, offering promising insights for future clinical cancer therapies.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Anbazhagan Sathiyaseelan
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Tieyan Jin
- College of Food Science and Engineering, Yanbian University, Yanji, Jilin 133002, China.
| | - Myeong-Hyeon Wang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
9
|
Kato R, Kamata Y, Tominaga M, Kishi R, Kaneko T, Tsujimura A, Suga Y, Takamori K. Possible Clinical Effects of Ketoconazole on Sorafenib-induced Hand-Foot Skin Reaction and Cytoprotection Mechanisms of Antifungal Agents against Multikinase Inhibitor-induced Keratinocyte Toxicity. Acta Derm Venereol 2025; 105:adv40697. [PMID: 40289816 PMCID: PMC12053523 DOI: 10.2340/actadv.v105.40697] [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: 05/02/2024] [Accepted: 03/13/2025] [Indexed: 04/30/2025] Open
Abstract
In recent years, molecular target drugs have become integral in treating malignant tumours. Multikinase inhibitors (MKIs) have been associated with serious skin disorders, including hand-foot skin reaction (HFSR), which impair patient quality of life, often disrupting activities of daily living necessitating dose reduction or discontinuation. As the pathogenic mechanisms of these skin disorders are unknown, no effective treatments have been established. Previously, by drug repurposing using an in vitro culture system, certain azole antifungal drugs (AFDs) were identified that prevented sorafenib-induced cell death of normal human epidermal keratinocytes. In this study, topical ketoconazole demonstrated clinical improvement in hyperkeratosis and pain associated with sorafenib-induced HFSR. Investigation of the mechanism using the in vitro culture system revealed sorafenib to be particularly cytotoxic among MKIs. Annexin V and TUNEL staining revealed apoptosis was mainly involved in this cytotoxicity. Antibody arrays and western blot showed increased levels of secretion of interleukin-1 receptor antagonist and macrophage migration inhibitory factor in culture supernatants. AFDs suppressed the secretion of these cytokines and reduced apoptosis in keratinocytes. This study reveals one aspect of the pathogenesis of sorafenib-induced HFSR and demonstrates that AFDs may be an effective treatment.
Collapse
Affiliation(s)
- Rui Kato
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan; Department of Dermatology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Yayoi Kamata
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Mitsutoshi Tominaga
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Ryoma Kishi
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan; Department of Dermatology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Takahide Kaneko
- Department of Dermatology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Akira Tsujimura
- Department of Urology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Yasushi Suga
- Department of Dermatology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Kenji Takamori
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan; Department of Dermatology, Juntendo University Urayasu Hospital, Chiba, Japan.
| |
Collapse
|
10
|
Zakariah M, Agishi G, Musa EZ, Dasa JJ, Majama YB, Gazali YA, Mahdy MAA. Rate of spermatogenic cell apoptosis in the testis of domestic chicken (Gallus gallus domesticus) at different age groups. Poult Sci 2025; 104:104953. [PMID: 40031382 PMCID: PMC11919428 DOI: 10.1016/j.psj.2025.104953] [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: 12/20/2024] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
In both normal and pathological conditions, excess spermatogenic cells in the testicular tissue are known to be removed by the mechanism of apoptosis. There are a few studies on spermatogenic cell apoptosis in avian species. Therefore, the purpose of this study was to evaluate the rate of spermatogenic cell apoptosis in three different age groups of the local domestic chicken (Gallus gallus domesticus). Samples were collected from the testes of domestic chicken at three different reproductive stages; pre-pubertal, pubertal, and adult. The samples were subjected to transmission electron microscopy (TEM) and flow cytometry evaluations. TEM results revealed the morphological characteristics of apoptotic cells which included irregular nuclear and plasma membranes in the early stages of apoptosis, nuclear membrane rupture, nuclear material condensation, and fragments of apoptotic bodies in the later stages of apoptosis. The flow cytometry results revealed a significant difference between the mean percentage of apoptotic spermatogenic cells for the three age groups (P < 0.05). Post hoc analysis revealed a significant difference in the adult age group relative to the pre-pubertal age group. However, there was no significant difference between apoptotic spermatogenic cells of the pre-pubertal and the pubertal, and between the pubertal and the adult age groups. In conclusion, the present study revealed a gradual increase in the rate of apoptotic spermatogenic cells in the testes of domestic chicken during the pre-pubertal, pubertal, and adult age groups.
Collapse
Affiliation(s)
- Musa Zakariah
- Department of Veterinary Anatomy, College of Veterinary Medicine, Federal University of Agriculture, P. M. B. 28, Zuru, Kebbi, Nigeria; Department of Veterinary Anatomy, Faculty of Veterinary Medicine, P. M. B. 1069 University of Maiduguri, Maiduguri, Nigeria.
| | - Geado Agishi
- Department of Veterinary Pathology, College of Veterinary Medicine, Federal University of Agriculture, P. M. B. 28, Zuru, Kebbi, Nigeria
| | - Esther Z Musa
- Department of Biological Science, College of Science, Federal University of Agriculture, P.M. B. 28, Zuru, Kebbi, Nigeria
| | - Josephine J Dasa
- Department of Biological Science, College of Science, Federal University of Agriculture, P.M. B. 28, Zuru, Kebbi, Nigeria
| | - Yagana B Majama
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, P. M. B. 1069 University of Maiduguri, Maiduguri, Nigeria
| | - Yagana A Gazali
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, P. M. B. 1069 University of Maiduguri, Maiduguri, Nigeria
| | - Mohammed A A Mahdy
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt; Department of Anatomy and Histology, Faculty of Veterinary Medicine, King Salman International University, Ras Sudr, Egypt
| |
Collapse
|
11
|
Hossain R, Kongchain A, Chatatikun M, Klangbud WK, Yupanqui CT, Majima HJ, Indo HP, Sompol P, Sekeroglu N, Phongphithakchai A, Tangpong J. Green Tea Pressurized Hot Water Extract in Atherosclerosis: A Multi-Approach Study on Cellular, Animal, and Molecular Mechanisms. Antioxidants (Basel) 2025; 14:404. [PMID: 40298660 PMCID: PMC12024429 DOI: 10.3390/antiox14040404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Atherosclerosis is a persistent inflammatory disorder influenced by oxidative stress and lipid imbalances, and it continues to be a major contributor to cardiovascular diseases. Rich in catechins and flavonoids, green tea pressurized hot water extract (GPHWE) demonstrated potent antioxidant activity through DPPH, ABTS, hydroxyl, and nitric oxide scavenging assays. In vitro, GPHWE protected RAW264.7 macrophages from oxidized LDL (Ox-LDL)-induced cytotoxicity and apoptosis by mitigating oxidative stress and enhancing cell survival. Animal studies using mice fed a high-fat diet (HFD) revealed notable improvements in lipid profiles, including decreases in total cholesterol, LDL, the atherosclerosis index (AI), the coronary risk index (CRI), and triglycerides, as well as lower levels of malondialdehyde (MDA), an indicator of oxidative stress. These results were comparable to those achieved with Simvastatin. Molecular docking studies indicated strong binding affinities of catechins to essential targets such as LOX-1, HMG-CoA reductase, caspase-3, and Nrf2, implying that the mechanisms of GPHWE involve antioxidant properties, regulation of lipids, and stabilization of plaques. The catechins of GPHWE, including epigallocatechin gallate (EGCG), epicatechin gallate (ECG), and epigallocatechin (EGC), were tentatively identified through qualitative analysis performed by UHPLC-QTOF-MS. This comprehensive approach positions GPHWE as a promising natural remedy for preventing atherosclerosis and reducing cardiovascular risk.
Collapse
Affiliation(s)
- Rahni Hossain
- College of Graduate Studies, Walailak University, Nakhon Si Thammarat 80160, Thailand;
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand; (A.K.); (M.C.); (H.J.M.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Anawat Kongchain
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand; (A.K.); (M.C.); (H.J.M.)
| | - Moragot Chatatikun
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand; (A.K.); (M.C.); (H.J.M.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Wiyada Kwanhian Klangbud
- Medical Technology Department, Faculty of Science, Nakhon Phanom University, Nakhon Phanom 48000, Thailand;
| | - Chutha Takahashi Yupanqui
- Center of Excellence in Functional Foods and Gastronomy, Faculty of Agro-Industry, Prince of Songkla University, Songkhla 90110, Thailand;
| | - Hideyuki J. Majima
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand; (A.K.); (M.C.); (H.J.M.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Hiroko P. Indo
- Department of Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima City 890-8544, Japan;
| | - Pradoldej Sompol
- Department of Pharmacology & Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Nazim Sekeroglu
- Department of Biology, Faculty of Arts and Sciences, Gaziantep University, 27310 Gaziantep, Turkey;
| | - Atthaphong Phongphithakchai
- Nephrology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand;
| | - Jitbanjong Tangpong
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand; (A.K.); (M.C.); (H.J.M.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
12
|
Teixeira T, Palmeira-Mello MV, Machado PH, Moraes CAF, Pinto C, Costa RC, Badaró W, Gomes Neto JA, Ellena J, Vieira Rocha F, Batista AA, Correa RS. Ru(II)-Fenamic-Based Complexes as Promising Human Ovarian Antitumor Agents: DNA Interaction, Cellular Uptake, and Three-Dimensional Spheroid Models. Inorg Chem 2025; 64:3707-3718. [PMID: 39962996 PMCID: PMC11884657 DOI: 10.1021/acs.inorgchem.4c04344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 03/04/2025]
Abstract
Cancer resistance to chemotherapeutic agents such as cisplatin presents a significant challenge, leading to treatment failure and poor outcomes. Novel metal-based compounds offer a promising strategy to overcome drug resistance and to enhance efficacy. Four Ru(II) complexes with fenamic acid derivatives were synthesized and characterized: [Ru(L)(bipy)(dppp)]PF6, where L represents fenamic acid (HFen, complex 1), mefenamic acid (HMFen, complex 2), tolfenamic acid (HTFen, complex 3), and flufenamic acid (HFFen, complex 4). Their composition was supported by molar conductivity, elemental analysis, Fourier transform infrared spectroscopy, ultraviolet-visible spectroscopy, mass spectrometry, and 31P{1H}, 1H, and 13C nuclear magnetic resonance, with the crystal structure of complex 1 confirmed via X-ray diffraction. Complexes 1-4 exhibited notable cytotoxicity against tested cell lines, particularly A2780 and A2780cisR (cisplatin-resistant ovarian tumors), compared to MDA-MB-231 (breast) and A549 (lung) lines. Mechanistic studies revealed weak DNA interactions through minor grooves or electrostatic binding. Cellular uptake assays showed effective internalization of complexes 1 (3.6%) and 2 (4.5%), correlating with potent IC50 values. These complexes also altered cell morphology, reduced cell density, and inhibited colony formation in the A2780 cells. Staining assays indicated induced cell death and organelle damage, highlighting their potential as promising antitumor agents.
Collapse
Affiliation(s)
- Tamara Teixeira
- Department
of Chemistry, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35402-136 Ouro
Preto, Minas Gerais, Brazil
- Department
of Chemistry, Federal University of São
Carlos (UFSCar), 13565-905 São Carlos, São Paulo, Brazil
| | - Marcos V. Palmeira-Mello
- Department
of Chemistry, Federal University of São
Carlos (UFSCar), 13565-905 São Carlos, São Paulo, Brazil
| | - Pedro Henrique Machado
- Department
of Chemistry, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35402-136 Ouro
Preto, Minas Gerais, Brazil
- Department
of Chemistry, Federal University of São
Carlos (UFSCar), 13565-905 São Carlos, São Paulo, Brazil
| | - Carlos A. F. Moraes
- Department
of Chemistry, Federal University of São
Carlos (UFSCar), 13565-905 São Carlos, São Paulo, Brazil
| | - Camila Pinto
- Institute
of Physics of São Carlos, University of São Paulo (IFSC/USP), 13566-590 São
Carlos, São Paulo, Brazil
| | - Rayane C. Costa
- Institute
of Chemistry, São Paulo State University (UNESP), 14800-900 Araraquara, São Paulo, Brazil
| | - Wladimir Badaró
- Institute
of Chemistry, São Paulo State University (UNESP), 14800-900 Araraquara, São Paulo, Brazil
| | - José A. Gomes Neto
- Institute
of Chemistry, São Paulo State University (UNESP), 14800-900 Araraquara, São Paulo, Brazil
| | - Javier Ellena
- Institute
of Physics of São Carlos, University of São Paulo (IFSC/USP), 13566-590 São
Carlos, São Paulo, Brazil
| | - Fillipe Vieira Rocha
- Department
of Chemistry, Federal University of São
Carlos (UFSCar), 13565-905 São Carlos, São Paulo, Brazil
| | - Alzir A. Batista
- Department
of Chemistry, Federal University of São
Carlos (UFSCar), 13565-905 São Carlos, São Paulo, Brazil
| | - Rodrigo S. Correa
- Department
of Chemistry, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35402-136 Ouro
Preto, Minas Gerais, Brazil
| |
Collapse
|
13
|
Sun S, Fan R, Chang L, Gao L, Liu C, Liu D, Niu S. Isoferulic Acid Inhibits Proliferation and Migration of Pancreatic Cancer Cells, and Promotes the Apoptosis of Pancreatic Cancer Cells in a Mitochondria-Dependent Manner Through Inhibiting NF-κB Signalling Pathway. Clin Exp Pharmacol Physiol 2025; 52:e70025. [PMID: 39861953 DOI: 10.1111/1440-1681.70025] [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: 06/21/2024] [Revised: 12/01/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Isoferulic acid (IA), a derivative of cinnamic acid, is derived from Danshen and exhibits anticancer properties by disrupting cancer cell activities. However, its role in pancreatic cancer, the "king of cancer", was unknown. In this study, pancreatic cancer cells were subjected to treatment with IA (6.25, 12.5, 25 μM), and nude mice injected with pancreatic cancer cells were received IA at doses of 7.5 mg/kg/day or 30 mg/kg/day by oral administration. CCK8, Annexin V-FITC/propidium iodide (PI) double staining and TUNEL assay were conducted to evaluate the cell viability and apoptosis. Hoechst staining and comet assay was employed to measure DNA damage. Mitochondrial membrane potential (MMP) analysis was carried out to explain the mitochondrial damage. EdU and wound healing assay were performed for cell proliferation and migration detection. Immunofluorescence and western blot were used to explore the mechanism. We found that IA reduced cell viability and induced apoptosis, as evidenced by an increase in Annexin V-FITC+PI- and Annexin V-FITC+PI+ cell populations, brighter TUNEL and Hoechst staining, and more percentage of tail DNA. Furthermore, IA decreased MMP and changed levels of apoptosis-related proteins. The cell proliferation and migration were inhibited by IA treatment. Mechanically, IA downregulated the phosphorylation of IĸBα and inhibited p65 nuclear translocation, consequently suppressing NF-κB pathway. In general, IA suppressed the cell proliferation and migration, and caused apoptosis of pancreatic cancer cells in a mitochondria-dependent manner through blocking NF-κB signalling pathway, indicating that IA may be a potential therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Suqin Sun
- First Department of Pulmonary Disease, Hanan Branch of the Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Rong Fan
- Department of Endocrinology, Hanan Branch of the Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Li Chang
- First Ward of Oncology Department, Harbin Chest Hospital, Harbin, People's Republic of China
| | - Lei Gao
- Department of Oncology, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Chunting Liu
- First Department of Pulmonary Disease, Hanan Branch of the Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Dongying Liu
- First Department of Pulmonary Disease, Hanan Branch of the Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Shiyu Niu
- Department of Endocrinology, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| |
Collapse
|
14
|
Kuldeep, Jena SR, Samanta L, Subuddhi U. Anthracene-Conjugated Steroidal Amphiphiles: Soft Functional Materials Exhibiting Supramolecular Aggregation Induced Enhanced Emission with Potential Applications as Drug Carriers and Fluorescent Bioprobes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:1157-1174. [PMID: 39745081 DOI: 10.1021/acs.langmuir.4c04502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Bile salts (BS) are naturally occurring steroidal biosurfactants. The ease of functionalization of BSs has boosted their use as inexpensive building blocks for the fabrication of a broad set of value-added soft functional materials. In the present work, three fluorescent bile acid (FBA) derivatives have been synthesized by conjugating anthracene at the side chain of lithocholic acid, deoxycholic acid, and cholic acid to understand the effect of the nature of the steroid nucleus on their physicochemical properties. In an aqueous medium, the FBAs showed a strong supramolecular aggregation propensity, even in the micromolar concentration range, which is in contrast to their BS analogues that form micelles mostly in the millimolar range. The FBA aggregation leads to a prearranged geometry in the ground state with a favorable orientation of anthracene units for excimer formation on excitation, leading to supramolecular aggregation-induced enhanced emission (AIEE). A detailed investigation reveals the pivotal role of the steroidal skeleton in their aggregation propensity and optical behavior. The FBA assemblies, with ordered structures plus anthracene being a part of their building blocks, are endowed with interesting properties different from those in dilute organic media, which makes them extremely attractive for diverse applications, e.g., as potential drug carriers owing to their ability to serve as efficient hosts for the protective encapsulation of hydrophobic guests; as membrane probes and bioimaging agents due to their efficient membrane permeability and cell-imaging ability; and as system probes because of their remarkable sensitivity toward the aggregation process of natural bile salts in the aqueous medium. Therefore, the present study not only enhances the fundamental understanding of this unique class of amphiphiles but also opens new prospects in tailoring novel self-assembled soft functional materials. Moreover, it offers a benchmark for developing BS-based fluorescent derivatives with unique photophysical characteristics for applications as potential bioprobes.
Collapse
Affiliation(s)
- Kuldeep
- Department of Chemistry, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Soumya Ranjan Jena
- Department of Zoology, Ravenshaw University, Cuttack, Odisha 753003, India
| | - Luna Samanta
- Department of Zoology, Ravenshaw University, Cuttack, Odisha 753003, India
| | - Usharani Subuddhi
- Department of Chemistry, National Institute of Technology, Rourkela, Odisha 769008, India
| |
Collapse
|
15
|
Dorairaj DP, Kumar P, Rajasekaran H, Bhuvanesh N, Hsu SCN, Karvembu R. Copper(II) complexes containing hydrazone and bipyridine/phenanthroline ligands for anticancer application against breast cancer cells. J Inorg Biochem 2025; 262:112759. [PMID: 39426333 DOI: 10.1016/j.jinorgbio.2024.112759] [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: 06/26/2024] [Revised: 09/29/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
In this work, mixed ligand Cu(II) complexes containing hydrazone and bipyridine ligands (CB1-CB5), or hydrazone and phenanthroline ligands (CP1-CP5) have been synthesized and characterized by spectroscopic and analytical techniques. Single crystal X-ray structure of complex CB1 revealed that two nitrogen atoms from bipyridine, one carbonyl oxygen, one azomethine nitrogen and one hydroxyl oxygen from the hydrazone ligand coordinated to Cu(II) ion, adopting a distorted square pyramidal geometry. Interaction of these complexes with calf thymus (CT) DNA and bovine serum albumin (BSA) was analyzed by absorption and emission studies. Further, the in vitro anticancer activity of the complexes was investigated exclusively against the breast cancer cells namely MCF7, T47D and MDA MB 231, and a normal breast MCF 10a cell line. The phenanthroline bearing complexes (CP1-CP5) displayed better activity than their bipyridine counterparts as seen from the IC50 values. In addition, the most active complex CP1 having an IC50 value of 5.8 ± 0.3 μM against T47D cancer cells was investigated for its mode of cell death through acridine orange/ethidium bromide(AO/EB), 4',6-diamidino-2-phenylindole (DAPI) and Annexin-V fluorescein isothiocyanate (FITC) staining assays which revealed apoptosis. Lastly, the cell cycle analysis revealed that complex CP1 induced cell death in T47D cancer cells at the G0/G1 phase.
Collapse
Affiliation(s)
| | - Prashant Kumar
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India
| | - Haritha Rajasekaran
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India
| | - Nattamai Bhuvanesh
- Department of Chemistry, Texas A & M University, College Station, TX 77842, USA
| | - Sodio C N Hsu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ramasamy Karvembu
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India.
| |
Collapse
|
16
|
Das S, Patel T, Himaja A, Regula S, Banerjee S, De AK, Qureshi IA, Gayen S, Ghosh B, Adhikari N, Jha T. Derivatives of D(-)-glutamine-based MMP-2 inhibitors as an effective remedy for the management of chronic myeloid leukemia-Part-III: Synthesis, biological screening and in silico binding interaction analysis. Bioorg Chem 2025; 154:108057. [PMID: 39708552 DOI: 10.1016/j.bioorg.2024.108057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Tyrosine kinase inhibitors (TKIs) have markedly improved the overall survival rate of patients with chronic myeloid leukemia (CML), enabling them to achieve a normal life expectancy. However, toxicity, relapse, and drug resistance continue to pose major challenges in the clinical treatment of CML. The progression of leukemia is directly connected to higher expression levels and enzymatic actions of matrix metalloproteinase-2 (MMP-2). It is also associated with increased expression and enzymatic actions of matrix metalloproteinase-9 (MMP-9). From this perspective, MMP-2 and MMP-9 offers a promising strategy for developing novel therapeutic molecules that could be effective in treating CML. This study is the Part-III of D(-)-glutamine-based MMP-2 inhibitors series for the management of chronic myeloid leukemia. Fourteen newly synthesized p-tosyl-D(-)-glutamine derivatives were examined in cell culture-based antileukemic assays and also evaluated for their ability to inhibit MMPs. The lead compounds 5g and 5j demonstrated the most promising antileukemic potential. Compounds 5g and 5j are safe for normal cells and effectively block gelatinases (MMP-2 and MMP-9). The best active molecule 5g induced significant apoptosis. Compound 5g reduced MMP-2 levels in the K562 cell line. It also had strong antiangiogenic effects in the ACHN cell line. The strongest MMP-2 inhibitor, 5g, had stable binding at the MMP-2 active site, which is linked to its effective inhibition of MMP-2. In conclusion, these p-tosyl-D(-)-glutamine derivatives are promising MMP-2 inhibitors. They have strong anti-CML effects and should be studied more for future CML treatment.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Matrix Metalloproteinase 2/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Matrix Metalloproteinase Inhibitors/pharmacology
- Matrix Metalloproteinase Inhibitors/chemistry
- Matrix Metalloproteinase Inhibitors/chemical synthesis
- Glutamine/chemistry
- Glutamine/metabolism
- Glutamine/pharmacology
- Structure-Activity Relationship
- Molecular Structure
- Drug Screening Assays, Antitumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Molecular Docking Simulation
Collapse
Affiliation(s)
- Sanjib Das
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India; School of Pharmacy, Sister Nivedita University, DG Block (New Town), Action Area 1, 1/2, Newtown, Chakpachuria, Kolkata 700156, India
| | - Tarun Patel
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Ambati Himaja
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Sanjeev Regula
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Asit Kumar De
- Department of Chemistry, Jadavpur University, Kolkata, India
| | - Insaf Ahmed Qureshi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500 046, Telangana, India
| | - Shovanlal Gayen
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India.
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
17
|
Spina A, Guidarelli A, Buffi G, Fiorani M, Cantoni O. Unveiling the link between NADPH oxidase 2 activation and mitochondrial superoxide formation in leukemic cell killing induced by arsenic trioxide. Pharmacol Res 2025; 211:107554. [PMID: 39694125 DOI: 10.1016/j.phrs.2024.107554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
This study focused on the interplay between NADPH oxidase 2 (NOX 2) activation and mitochondrial superoxide (mitoO2.-) formation induced by clinically relevant concentrations of arsenic trioxide (ATO; As2O3) in acute promyelocytic leukemia (APL) cells. Carefully controlled inhibitor studies and small interfering RNA mediated downregulation of p47phox (a component of the NOX 2 complex) expression demonstrated that, in an APL cell line, ATO promotes upstream NOX 2 activation critically connected with the formation of mitoO2.- and with the ensuing mitochondrial permeability transition (MPT)-dependent apoptosis. Instead, acute myeloid leukemia (AML) cell lines respond to ATO with low NOX 2 activation, resulting in a state that is non-permissive for mitoO2.- formation. Consistently, through rescue experiments, we demonstrate that pharmacological stimulation of NOX 2 overcomes resistance in these cells, thereby initiating the same cascade of downstream events observed in APL cells. As a final note, several lines of evidence, including measurement of glutathione, catalase and glutathione peroxidase levels, indicated that the antioxidant machinery was similar in APL and AML cells. The results regarding nuclear factor erythroid 2 p45-related factor 2-dependent antioxidant responses were instead of more complex interpretation as NB4 cells appeared particularly responsive to ATO. Our findings allow a novel interpretation of the interplay between NOX 2 activation and mitoO2.- formation induced by ATO, ultimately steering leukemic cells towards MPT-dependent apoptosis. These mechanistic insights provide a rationale for the disparate responses of APL and AML cells to ATO, offering potential avenues for the development of therapeutic intervention tailored to specific leukemia subtypes.
Collapse
MESH Headings
- Arsenic Trioxide/pharmacology
- Humans
- Superoxides/metabolism
- Mitochondria/drug effects
- Mitochondria/metabolism
- Antineoplastic Agents/pharmacology
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/pathology
- NADPH Oxidase 2/metabolism
- NADPH Oxidase 2/genetics
- Arsenicals/pharmacology
- Cell Line, Tumor
- Oxides/pharmacology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Apoptosis/drug effects
- Enzyme Activation/drug effects
- NADPH Oxidases/metabolism
- NADPH Oxidases/genetics
Collapse
Affiliation(s)
- Andrea Spina
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Gloria Buffi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mara Fiorani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
18
|
Dias C, Lourenço CF, Laranjinha J, Ledo A. High-Resolution Respirometry Methodology for Bioenergetic and Metabolic Studies in Intact Brain Slices. Methods Mol Biol 2025; 2878:35-48. [PMID: 39546255 DOI: 10.1007/978-1-0716-4264-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
The brain is critically dependent on energetic substrates as it consumes circa 20% of glucose and oxygen under normal physiological conditions. Although different cell types and at different locations might experience particular specificities in the utilization of these substrates, overall, mitochondrial oxidative phosphorylation supports the most efficient energy transduction process, enabling the complete oxidation of glucose to CO2 coupled to ATP synthesis in the presence of O2. Impairment of mitochondrial bioenergetics has been identified as an early event in many brain diseases and aging. Thus, novel methodologies to readily assess mitochondrial respiration in brain tissue, while preserving cellular and mitochondrial architecture and overcoming the serious drawbacks of studies using isolated mitochondrial preparations, are needed. Here we describe a methodology for studying functional parameters defining tissue metabolic respiration in brain hippocampal slices. The methodology can be used for physiological, pharmacological, and toxicological studies.
Collapse
Affiliation(s)
- Cândida Dias
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Cátia F Lourenço
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - João Laranjinha
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ana Ledo
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
19
|
Izadifar M, Massumi M, Prentice KJ, Oussenko T, Li B, Elbaz J, Puri M, Wheeler MB, Nagy A. Microfluidic chip systems for characterizing glucose-responsive insulin-secreting cells equipped with FailSafe kill-switch. Stem Cell Res Ther 2024; 15:486. [PMID: 39696686 DOI: 10.1186/s13287-024-04059-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/10/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Pluripotent cell-derived islet replacement therapy offers promise for treating Type 1 diabetes (T1D), but concerns about uncontrolled cell proliferation and tumorigenicity present significant safety challenges. To address the safety concern, this study aims to establish a proof-of-concept for a glucose-responsive, insulin-secreting cell line integrated with a built-in FailSafe kill-switch. METHOD We generated β cell-induced progenitor-like cells (βiPLCs) from primary mouse pancreatic β cells through interrupted reprogramming. Then, we transcriptionally linked our FailSafe (FS) kill-switch, HSV-thymidine kinase (TK), to Cdk1 gene using a CRISPR/Cas9 knock-in strategy, resulting in a FailSafe βiPLC line, designated as FSβiPLCs. Subsequently we evaluated and confirmed the functionality of the drug-inducible kill-switch in FSβiPLCs at different ganciclovir (GCV) concentrations using our PDMS-based transcapillary microfluidic system. Finally, we assessed the functionality of FSβiPLCs by characterizing the dynamics of insulin secretion in response to changes in glucose concentration using our microfluidic perfusion glucose-stimulated insulin secretion (GSIS) assay-on- chip. RESULTS The βiPLCs exhibited Ins1, Pdx1 and Nkx6.1 expression, and glucose responsive insulin secretion, the essential properties of pancreatic beta cells. The βiPLCs were amenable to genome editing which allowed for the insertion of the kill-switch into the 3'UTR of Cdk1, confirmed by PCR genotyping. Our transcapillary microfluidic system confirmed the functionality of the drug-inducible kill-switch in FSβiPLCs, showing an effective cell ablation of dividing cells from a heterogeneous cell population at different ganciclovir (GCV) concentrations. The Ki67 expression assessment further confirmed that slow- or non-dividing cells in the FSβiPLC population were resistant to GCV. Our perfusion glucose-stimulated insulin secretion (GSIS) assay-on-chip revealed that the resistant non-dividing FSβiPLCs exhibited higher levels of insulin secretion and glucose responsiveness compared to their proliferating counterparts. CONCLUSIONS This study establishes a proof-of-concept for the integration of a FailSafe kill-switch system into a glucose-responsive, insulin-secreting cell line to address the safety concerns in stem cell-derived cell replacement treatment for T1D. The microfluidic systems provided valuable insights into the functionality and safety of these engineered cells, demonstrating the potential of the kill-switch to reduce the risk of tumorigenicity in pluripotent cell-derived insulin-secreting cells.
Collapse
Affiliation(s)
- Mohammad Izadifar
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Mohammad Massumi
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Kacey J Prentice
- Departments of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tatiana Oussenko
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Biao Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Judith Elbaz
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Mira Puri
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Michael B Wheeler
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
20
|
Polechońska L, Klink A, Sokołowska K. Rising temperature impacts the trace metal uptake and toxicity in aquatic plants - A case study of Ni and Co in Elodea canadensis Michx. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:177307. [PMID: 39481559 DOI: 10.1016/j.scitotenv.2024.177307] [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: 08/09/2024] [Revised: 10/09/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
The global warming and environmental pollution are two crucial contemporary concerns. As both are strongly connected with urbanisation and anthropogenic impact on the environment, they often affect the ecosystem simultaneously. Aquatic habitats are particularly susceptible to thermal and chemical pollution. Temperature influences nearly all physical and chemical features of water bodies and trace metals are known for their toxicity to aquatic organisms. However, effects of multiple stressors, cumulative effects as well as response and possible adaptations of organisms are still open questions. Thus, the aim of this study was to characterize the combined effect of temperature and two trace metals (Co and Ni) on the metal bioaccumulation and viability of a model aquatic macrophyte Elodea canadensis Michx. We exposed shoots of E. canadensis to three temperatures and four metal concentrations (together and separately) applied at environmentally relevant levels. Shoot growth and metal concentration in plants were measured after 120 h. Moreover, after 24, 72 and 120 h the changes in leaf cell morphology and viability were analysed. The results showed that metal accumulation was dose-dependent and was not affected by temperature. The growth of plants was not affected by temperature nor metals. On the other hand, the exposure to Co and Ni and the elevated temperature negatively affected cell viability of E. canadensis leaves which manifested by increased permeability of plasma membranes and visible necroses. The greatest damaged leaf areas were determined after 120 h in the highest concentration of both metals and the highest temperature which indicates synergistic impact of trace metals and temperature on performance of macrohydrophytes. The observed phenomena suggest that global warming and/or thermal pollution may have implications for the performance of aquatic macrophytes in chemically polluted waters, their ability to spread and colonize polluted habitats and their suitability in phytoremediation.
Collapse
Affiliation(s)
- Ludmiła Polechońska
- University of Wrocław, Faculty of Biological Sciences, Department of Ecology, Biogeochemistry and Environmental Protection, Kanonia 6/8, 50-328 Wrocław, Poland.
| | - Agnieszka Klink
- University of Wrocław, Faculty of Biological Sciences, Department of Ecology, Biogeochemistry and Environmental Protection, Kanonia 6/8, 50-328 Wrocław, Poland
| | - Katarzyna Sokołowska
- University of Wrocław, Faculty of Biological Sciences, Department of Plant Developmental Biology, Kanonia 6/8, Wrocław 50-328, Poland
| |
Collapse
|
21
|
Shieu MK, Lin CC, Ho HY, Lo YS, Chuang YC, Hsieh MJ. Picrasidine I Regulates Apoptosis in Melanoma Cell Lines by Activating ERK and JNK Pathways and Suppressing AKT Signaling. ENVIRONMENTAL TOXICOLOGY 2024; 39:5309-5320. [PMID: 39194337 DOI: 10.1002/tox.24404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/21/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024]
Abstract
World Health Organization data indicate a continuous increase in melanoma incidence, with metastatic melanoma characterized by poor prognosis and drug resistance. The exploration of therapeutics derived from natural products remains an active area of in vitro research. The aim of this study was to determine the antitumor effects of picrasidine I, a natural compound extracted from Picrasma quassioides, against two melanoma cell lines. We selected two metastatic melanoma cell lines, HMY-1 and A2058, for molecular studies, including Western blotting, 4',6-diamidino-2-phenylindole staining, and flow cytometry. Picrasidine I demonstrated cytotoxic effects against the HMY-1 and A2058 melanoma cell lines. It induced cell cycle arrest in the sub-G1 phase and downregulated cell cycle-related proteins (e.g., cyclin A2, D1, cyclin-dependent kinases 4, and 6). In the intrinsic apoptosis pathway, picrasidine I activated proapoptotic proteins (e.g., Bax, Bak, t-Bid, BimL/S) and suppressed the expression of antiapoptotic proteins (e.g., Bcl-2, Bcl-xL), with an observed increase in the quantity of depolarized cells. In addition, the apoptotic effects of picrasidine I were linked to the activation of the c-Jun N-terminal kinase and extracellular signal-regulated kinase pathways and the inhibition of the protein kinase B signaling pathway. A human apoptosis array indicated claspin inhibition upon picrasidine I treatment, suggesting the potential involvement of picrasidine I in apoptosis and cell cycle regulation. Our findings suggest that picrasidine I has potential as a candidate for treating advanced melanoma, and thus these findings warrant further investigation. The modulation of claspin expression by picrasidine I could be investigated further as a potential biomarker to predict its efficacy in related to advanced stages of melanoma.
Collapse
Affiliation(s)
- Mu-Kuei Shieu
- Department of Dermatology, Changhua Christian Hospital, Changhua, Taiwan
| | - Chia-Chieh Lin
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsin-Yu Ho
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Yu-Sheng Lo
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Yi-Ching Chuang
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Ming-Ju Hsieh
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
22
|
Banach K, Kowalska J, Maszczyk M, Rzepka Z, Rok J, Wrześniok D. An In Vitro Strategy to Evaluate Ketoprofen Phototoxicity at the Molecular and Cellular Levels. Int J Mol Sci 2024; 25:12647. [PMID: 39684359 DOI: 10.3390/ijms252312647] [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: 10/17/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 12/18/2024] Open
Abstract
Phototoxicity is a significant problem that occurs in a large part of the population and is often caused by commonly used pharmaceuticals, including over-the-counter drugs. Therefore, testing drugs with photosensitizing potential is very important. The aim of this study is to analyze the cytotoxicity and phototoxicity of ketoprofen towards human melanocytes and fibroblasts in three different treatment schemes in order to optimize the study. Cytometric tests (studies of viability, proliferation, intracellular thiol levels, mitochondrial potential, cell cycle, and DNA fragmentation), Western blot analysis (cytochrome c and p44/p42 protein levels), and confocal microscopy imaging were performed to assess the impact of the developed treatments on skin cells. Research on experimental schemes may help reduce or eliminate the risk of phototoxic reactions. In the case of ketoprofen, we found that the strongest phototoxic potential was exhibited in the treatment where the drug was present in the solution during the irradiation of cells, both pigmented and non-pigmented cells. These results indicate that the greatest risk of photosensitivity reactions related to ketoprofen occurs after direct contact with the drug and UV exposure.
Collapse
Affiliation(s)
- Klaudia Banach
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Justyna Kowalska
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Mateusz Maszczyk
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Zuzanna Rzepka
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Jakub Rok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
23
|
Zanrè V, Bellinato F, Cardile A, Passarini C, Di Bella S, Menegazzi M. BRAF-Mutated Melanoma Cell Lines Develop Distinct Molecular Signatures After Prolonged Exposure to AZ628 or Dabrafenib: Potential Benefits of the Antiretroviral Treatments Cabotegravir or Doravirine on BRAF-Inhibitor-Resistant Cells. Int J Mol Sci 2024; 25:11939. [PMID: 39596009 PMCID: PMC11593403 DOI: 10.3390/ijms252211939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Melanoma is an aggressive cancer characterized by rapid growth, early metastasis, and poor prognosis, with resistance to current therapies being a significant issue. BRAF mutations drive uncontrolled cell division by activating the MAPK pathway. In this study, A375 and FO-1, BRAF-mutated melanoma cell lines, were treated for 4-5 months with RAF inhibitor dabrafenib or AZ628, leading to drug resistance over time. The resistant cells showed altered molecular signatures, with differences in cell cycle regulation and the propensity of cell death. Dabrafenib-resistant cells maintained high proliferative activity, while AZ628-resistant cells, especially A375 cells, exhibited slow-cycling, and a senescent-like phenotype with high susceptibility to ferroptosis, a form of cell death driven by iron. Antiretroviral drugs doravirine and cabotegravir, known for their effects on human endogenous retroviruses, were tested for their impact on these resistant melanoma cells. Both drugs reduced cell viability and colony formation in resistant cell lines. Doravirine was particularly effective in reactivating apoptosis and reducing cell growth in highly proliferative resistant cells by increasing tumor-suppressor proteins p16Ink4a and p27Kip1. These findings suggest that antiretroviral drugs can influence apoptosis and cell proliferation in RAF-inhibitor-resistant melanoma cells, offering potential therapeutic strategies for overcoming drug resistance.
Collapse
Affiliation(s)
- Valentina Zanrè
- Section of Biochemistry, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (V.Z.)
| | - Francesco Bellinato
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Piazzale Stefani 1, 37126 Verona, Italy;
| | - Alessia Cardile
- Section of Biochemistry, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (V.Z.)
| | - Carlotta Passarini
- Section of Biochemistry, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (V.Z.)
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy;
| | - Marta Menegazzi
- Section of Biochemistry, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (V.Z.)
| |
Collapse
|
24
|
Hasan Mujahid M, Upadhyay TK, Upadhye V, Sharangi AB, Saeed M. Phytocompound identification of aqueous Zingiber officinale rhizome (ZOME) extract reveals antiproliferative and reactive oxygen species mediated apoptotic induction within cervical cancer cells: an in vitro and in silico approach. J Biomol Struct Dyn 2024; 42:8733-8760. [PMID: 37639378 DOI: 10.1080/07391102.2023.2247089] [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: 06/20/2023] [Accepted: 08/07/2023] [Indexed: 08/31/2023]
Abstract
The prevalence of cervical cancer in women is in the fourth position among various other types of cancer globally. Many established therapies, including surgery, chemotherapy, and immunomodulation, are present, but high levels of side effects cause mortality and morbidity among the patients. Zingiber officinale rhizome (ZOME) has been potentially used to cure a variety of ailments and diseases. The aqueous ZOME extract also contains ample phytochemical constituents having anticancer effects on different cancers. The cell viability of HeLa cells was evaluated using MTT assay with IC50 at 97 µg/mL. Furthermore, a significant level of ROS generation causes the apoptosis of the cells. Nuclear staining dye DAPI and Hoechst 33342 showed DNA's fragmented and condensed form. Propidium Iodide staining showed necrotic or late-apoptotic cells. While acidic organelle dye LysoTracker and MitoTracker dye along with dual staining showed significant results. In silico studies were carried out using identified phytochemicals from GC-MS analysis with pharmacokinetics properties (ADMET), and targeted toward receptor proteins for molecular docking. Ligands with high docked scores were subjected to molecular dynamics simulations at 310 K for 100 ns. In vitro and in silico investigations in our studies showed that aqueous ZOME extract can be used as an efficient therapy against cervical cancer treatment as it showed significant cytotoxic and antiproliferative effects toward the HeLa cell line.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mohd Hasan Mujahid
- Department of Biotechnology, Parul Institute of Applied Sciences and Research and Development Cell, Parul University, Vadodara, Gujarat, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Research and Development Cell, Parul University, Vadodara, Gujarat, India
| | - Vijay Upadhye
- Department of Biotechnology, Parul Institute of Applied Sciences and Research and Development Cell, Parul University, Vadodara, Gujarat, India
| | - Amit Baran Sharangi
- Department of Plantation, Spices, Medicinal & Aromatic Crops, BCKV-Agricultural University, Mohanpur, West Bengal, India
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| |
Collapse
|
25
|
Arora S, Singh S, Mittal A, Desai N, Khatri DK, Gugulothu D, Lather V, Pandita D, Vora LK. Spheroids in cancer research: Recent advances and opportunities. J Drug Deliv Sci Technol 2024; 100:106033. [DOI: 10.1016/j.jddst.2024.106033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
|
26
|
Chatnarin S, Thirabunyanon M. Potential of β-D-glucan polysaccharide from Ophiocordyceps sinensis OS8 cultivated mycelium on anticancer activity via inducing liver cancer cell death apoptosis. Process Biochem 2024; 145:243-249. [DOI: 10.1016/j.procbio.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
27
|
Qayoom H, Mir MA. Mutant P53 modulation by cryptolepine through cell cycle arrest and apoptosis in triple negative breast cancer. Biomed Pharmacother 2024; 179:117351. [PMID: 39216450 DOI: 10.1016/j.biopha.2024.117351] [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: 07/21/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Triple Negative Breast cancer is an aggressive breast cancer subtype. It has a more aggressive clinical course, an earlier age of onset, a larger propensity for metastasis, and worse clinical outcomes as evidenced by a higher risk of recurrence and a shorter survival rate. Currently, the primary options for TNBC treatment are surgery, radiation, and chemotherapy. These treatments however remain ineffective due to recurrence. However, given that p53 mutations have been identified in more than 60-88 % of TNBC, translating p53 into the clinical situation is particularly important in TNBC. In this study, we screened and evaluated the therapeutic potential of cryptolepine (CRP) in TNBC in-vitro models being an anti-malarial drug it could be repurposed as an anti-cancer therapeutic targeting TNBC. Moreover, the cytotoxicity activity of cryptolepine to TNBC cells and a detailed anti-tumor mechanism in mutant P53 has not been reported before. METHODS MTT assays were used to examine the cytotoxicity and cell viability activity of Cryptolepine in TNBC, non-TNBC T47D and MCF-7 and non-malignant MCF10A cells. Scratch wound and clonogenic assay was used to evaluate the cryptolepine's effect on migration and colony forming ability of TNBC cells. Flow cytometry, MMP and DAPI was used to assess cell cycle arrest and cell apoptosis mechanism. The expression of proteins was detected by western blots. The differential expression of RNAs was evaluated by RT-PCR and the interaction between P53 and drug was evaluated computationally using in-silico approach and in-vitro using ChIP assay. RESULTS In this study, we found that cryptolepine has more preferential cytotoxicity in TNBC than non-TNBC cells. Notably, our studies revealed the mechanism by which cryptolepine induces intrinsic apoptosis and inhibit migration, colony formation ability, induce cell cycle arrest by inducing conformational change in the mutant P53 thereby increasing its DNA binding ability, hence activating its tumor suppressing potential significantly. CONCLUSION Our study revealed that CRP significantly reduced the proliferation, migration and colony forming ability of TNBC cells lines. Moreover, it was revealed that CRP induces cell cycle arrest and apoptosis by activating mutant P53 and enhancing its DNA binding ability to induce its tumor suppressing ability.
Collapse
Affiliation(s)
- Hina Qayoom
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Manzoor A Mir
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India.
| |
Collapse
|
28
|
Sharma V, Sinha ES, Singh J. Investigation of In Vitro Anti-cancer and Apoptotic Potential of Onion-Derived Nanovesicles Against Prostate and Cervical Cancer Cell Lines. Appl Biochem Biotechnol 2024; 196:6957-6973. [PMID: 38441782 DOI: 10.1007/s12010-024-04872-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 11/21/2024]
Abstract
Plant-derived compounds have recently garnered significant interest in the field of medicine due to their rich repertoire of phytochemicals, which holds promise for exploring novel therapies to treat cancer. This study embarks on the first-time investigation of the anti-cancerous effect of onion-derived nanovesicles (ODNVs). ODNVs were isolated employing differential centrifugation followed by ultracentrifugation and subsequent characterization using dynamic light scattering (DLS), field emission scanning electron microscopy (FESEM), and Fourier transform infrared spectroscopy (FTIR). Furthermore, we delineated the anti-cancerous effect of ODNVs on two cancer cell line models HeLa (cervical cancer) and PC-3 (prostate cancer) using MTT assay, DAPI-based DNA damage using immunofluorescence microscopy, colony formation assay, migration assay, cell cycle analysis, and evaluation of apoptosis using flow cytometry and western blotting. The findings revealed dose- and time-dependent anti-proliferative effects of ODNVs on both HeLa and PC3 cell lines, accompanied by selective cytotoxicity against cancer cells. Additional results highlighted that ODNVs prevented colony growth and induced S-phase cell cycle arrest. Apoptosis induction was evaluated through alterations in nuclear morphology and the number of apoptotic cells, which increased significantly after ODNV treatment in both cancer cell lines. Furthermore, annexin V/PI staining evaluation of apoptotic cells by flow cytometry demonstrated that ODNV treatment significantly increased the number of apoptotic cells in both PC-3 and HeLa cells. Finally, Western blot analysis indicated changes in apoptosis-related proteins including bcl-2, bax, and caspase-3, emphasizing that the anti-cancerous effect of ODNVs is attributed to the induction of apoptosis and suggests the unexplored anti-cancerous potential of ODNVs.
Collapse
Affiliation(s)
- Vinayak Sharma
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India
| | - Eshu Singhal Sinha
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India
| | - Jagtar Singh
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
29
|
Sasikumar J, Laha S, Naik B, Das SP. Enhanced visualization of nuclear staining and cell cycle analysis for the human commensal Malassezia. Sci Rep 2024; 14:20936. [PMID: 39251619 PMCID: PMC11385215 DOI: 10.1038/s41598-024-69024-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/30/2024] [Indexed: 09/11/2024] Open
Abstract
Malassezia is a lipophilic commensal yeast that resides mainly on the mammalian skin and is also found to associate with the internal organs. Dysbiosis of Malassezia is related to several diseases and often escapes detection as it is difficult to culture and maintain. Malassezia cell wall differs from other budding yeasts like S. cerevisiae due to the difference in the lipid content and is difficult to transform. In this study, we present a methodology to stain Malassezia's nucleus and perform cell cycle studies. However, staining presents a challenge due to its exceptionally thick cell wall with high lipid content, hindering conventional methods. Our novel methodology addresses this challenge and enables the staining of the Malassezia nucleus with a low background. This would allow researchers to visualize the overall nuclear health specifically nuclear morphology and analyze DNA content, crucial for cell cycle progression. By employing DNA-specific dyes like DAPI or Hoechst, we can observe the nuclear structure, and using PI we can differentiate cells in distinct cell cycle phases using techniques like flow cytometry. This novel staining methodology unlocks the door for in-depth cell cycle analysis in Malassezia which has challenged us through ages being refractory to genetic manipulations, paving the way for a deeper understanding of this commensal fungus and its potential role in human health.
Collapse
Affiliation(s)
- Jayaprakash Sasikumar
- Cell Biology and Molecular Genetics, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Suparna Laha
- Cell Biology and Molecular Genetics, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Bharati Naik
- Cell Biology and Molecular Genetics, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Shankar Prasad Das
- Cell Biology and Molecular Genetics, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India.
| |
Collapse
|
30
|
Myeza N, Slabber C, Munro OQ, Sookai S, Zacharias SC, Martins-Furness C, Harmse L. An 8-aminoquinoline-naphthyl copper complex causes apoptotic cell death by modulating the expression of apoptotic regulatory proteins in breast cancer cells. Eur J Pharmacol 2024; 978:176764. [PMID: 38908670 DOI: 10.1016/j.ejphar.2024.176764] [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: 02/27/2024] [Revised: 06/08/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Breast cancer is one of the most common cancers globally and a leading cause of cancer-related deaths among women. Despite the combination of chemotherapy with targeted therapy, including monoclonal antibodies and kinase inhibitors, drug resistance and treatment failure remain a common occurrence. Copper, complexed to various organic ligands, has gained attention as potential chemotherapeutic agents due to its perceived decreased toxicity to normal cells. The cytotoxic efficacy and the mechanism of cell death of an 8-aminoquinoline-naphthyl copper complex (Cu8AqN) in MCF-7 and MDA-MB-231 breast cancer cell lines was investigated. The complex inhibited the growth of MCF-7 and MDA-MB-231 cells with IC50 values of 2.54 ± 0.69 μM and 3.31 ± 0.06 μM, respectively. Nuclear fragmentation, annexin V binding, and increased caspase-3/7 activity indicated apoptotic cell death. The loss of mitochondrial membrane potential, an increase in caspase-9 activity, the absence of active caspase-8 and a decrease of tumour necrosis factor receptor 1(TNFR1) expression supported activation of the intrinsic apoptotic pathway. Increased ROS formation and increased expression of haem oxygenase-1 (HMOX-1) indicated activation of cellular stress pathways. Expression of p21 protein in the nuclei was increased indicating cell cycle arrest, whilst the expression of inhibitor of apoptosis proteins (IAPs); cIAP1, XIAP and survivin were decreased, creating a pro-apoptotic environment. Phosphorylated p53 species; phospho-p53(S15), phospho-p53(S46), and phospho-p53(S392) accumulated in MCF-7 cells indicating the potential of Cu8AqN to restore p53 function in the cells. In combination, the data indicates that Cu8AqN is a useful lead molecule worthy of further exploration as a potential anti-cancer drug.
Collapse
Affiliation(s)
- Nonzuzo Myeza
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, South Africa
| | - Cathy Slabber
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg, 2017, South Africa
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg, 2017, South Africa; School of Chemistry, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| | - Sheldon Sookai
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg, 2017, South Africa
| | - Savannah C Zacharias
- School of Chemistry and Physics, University of KwaZulu-Natal, King Edward Drive, Pietermaritzburg, Scottsville, 3209, South Africa
| | - Carla Martins-Furness
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, South Africa
| | - Leonie Harmse
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, South Africa.
| |
Collapse
|
31
|
Choudhury S, Mehra A, Srivastava S, Sharma M, Singh M, Panda JJ. Lipopolysaccharide targeting-peptide-capped chitosan gold nanoparticles for laser-induced antibacterial activity. Nanomedicine (Lond) 2024; 19:1913-1929. [PMID: 39225175 PMCID: PMC11457656 DOI: 10.1080/17435889.2024.2382073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Aim: We present the synthesis of anti-bacterial gold nanoparticles using chitosan as a dual-functional agent. The resulting ChAuNPs were further modified with a lipopolysaccharide-targeting antibacterial peptide to aid in biocompatibility and specificity.Materials & methods: The nanoparticles' antibacterial activity against Escherichia coli was tested in the presence of a 450 nm laser.Results: Our data suggested that the peptide and laser emissions had a synergistic impact on the gold nanoparticles, resulting in strong antibacterial effects. The study shows that advanced nanomaterials, including chitosan, gold nanoparticles and lipopolysaccharide targeting peptides, can boost antibacterial functions at a low concentration of 250 μg/ml.Conclusion: The findings highlight ChAuNPs' potential as strong antibacterial agents, with targeted alterations critical for maximizing their utilization.
Collapse
Affiliation(s)
| | - Aakrati Mehra
- Institute of Nano Sciences & Technology, Mohali, Punjab, 140306, India
| | | | - Manju Sharma
- Institute of Nano Sciences & Technology, Mohali, Punjab, 140306, India
| | - Manish Singh
- Institute of Nano Sciences & Technology, Mohali, Punjab, 140306, India
| | - Jiban Jyoti Panda
- Institute of Nano Sciences & Technology, Mohali, Punjab, 140306, India
| |
Collapse
|
32
|
Baidya SK, Patel T, Himaja A, Banerjee S, Das S, Ghosh B, Jha T, Adhikari N. Biphenylsulfonamides as effective MMP-2 inhibitors with promising antileukemic efficacy: Synthesis, in vitro biological evaluation, molecular docking, and MD simulation analysis. Drug Dev Res 2024; 85:e22255. [PMID: 39233391 DOI: 10.1002/ddr.22255] [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: 05/21/2024] [Revised: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
Overexpression of matrix metalloproteinase-2 (MMP-2) possesses a correlation with leukemia especially chronic myeloid leukemia (CML). However, no such MMP-2 inhibitor has come out in the market to date for treating leukemia. In this study, synthesis, biological evaluation, and molecular modeling studies of a set of biphenylsulfonamide derivatives as promising MMP-2 inhibitors were performed, focusing on their potential applications as antileukemic therapeutics. Compounds DH-18 and DH-19 exerted the most effective MMP-2 inhibition (IC50 of 139.45 nM and 115.16 nM, respectively) with potent antileukemic efficacy against the CML cell line K562 (IC50 of 0.338 µM and 0.398 µM, respectively). The lead molecules DH-18 and DH-19 reduced the MMP-2 expression by 21.3% and 17.8%, respectively with effective apoptotic induction (45.4% and 39.8%, respectively) in the K562 cell line. Moreover, both these compounds significantly arrested different phases of the cell cycle. Again, both these molecules depicted promising antiangiogenic efficacy in the ACHN cell line. Nevertheless, the molecular docking and molecular dynamics (MD) simulation studies revealed that DH-18 formed strong bidentate chelation with the catalytic Zn2+ ion through the hydroxamate zinc binding group (ZBG). Apart from that, the MD simulation study also disclosed stable binding interactions of DH-18 and MMP-2 along with crucial interactions with active site amino acid residues namely His120, Glu121, His124, His130, Pro140, and Tyr142. In a nutshell, this study highlighted the importance of biphenylsulfonamide-based novel and promising MMP-2 inhibitors to open up a new avenue for potential therapy against CML.
Collapse
Affiliation(s)
- Sandip K Baidya
- Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
- School of Pharmacy, Sister Nivedita University, Kolkata, India
| | - Tarun Patel
- Department of Pharmacy, Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Ambati Himaja
- Department of Pharmacy, Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Suvankar Banerjee
- Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
| | - Sanjib Das
- Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
- School of Pharmacy, Sister Nivedita University, Kolkata, India
| | - Balaram Ghosh
- Department of Pharmacy, Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Tarun Jha
- Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
| | - Nilanjan Adhikari
- Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
| |
Collapse
|
33
|
Das S, Mondal S, Patel T, Himaja A, Adhikari N, Banerjee S, Baidya SK, De AK, Gayen S, Ghosh B, Jha T. Derivatives of D(-) glutamine-based MMP-2 inhibitors as an effective remedy for the management of chronic myeloid leukemia-Part-I: Synthesis, biological screening and in silico binding interaction analysis. Eur J Med Chem 2024; 274:116563. [PMID: 38843586 DOI: 10.1016/j.ejmech.2024.116563] [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: 02/01/2024] [Revised: 04/23/2024] [Accepted: 05/31/2024] [Indexed: 06/17/2024]
Abstract
Chronic myeloid leukemia (CML) is a global issue and the available drugs such as tyrosine kinase inhibitors (TKIs) comprise various toxic effects as well as resistance and cross-resistance. Therefore, novel molecules targeting specific enzymes may unravel a new direction in antileukemic drug discovery. In this context, targeting gelatinases (MMP-2 and MMP-9) can be an alternative option for the development of novel molecules effective against CML. In this article, some D(-)glutamine derivatives were synthesized and evaluated through cell-based antileukemic assays and tested against gelatinases. The lead compounds, i.e., benzyl analogs exerted the most promising antileukemic potential showing nontoxicity in normal cell line including efficacious gelatinase inhibition. Both these lead molecules yielded effective apoptosis and displayed marked reductions in MMP-2 expression in the K562 cell line. Not only that, but both of them also revealed effective antiangiogenic efficacy. Importantly, the most potent MMP-2 inhibitor, i.e., benzyl derivative of p-tosyl D(-)glutamine disclosed stable binding interaction at the MMP-2 active site correlating with the highly effective MMP-2 inhibitory activity. Therefore, such D(-)glutamine derivatives might be explored further as promising MMP-2 inhibitors with efficacious antileukemic profiles for the treatment of CML in the future.
Collapse
Affiliation(s)
- Sanjib Das
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Subha Mondal
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Tarun Patel
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Ambati Himaja
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Sandip Kumar Baidya
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Asit Kumar De
- Department of Chemistry, Jadavpur University, Kolkata, India
| | - Shovanlal Gayen
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India.
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| |
Collapse
|
34
|
Poblano-Bata J, Zaragoza-Ojeda M, De Vizcaya-Ruiz A, Arenas-Huertero F, Amador-Muñoz O. Toxicological effects of solvent-extracted organic matter associated with PM 2.5 on human bronchial epithelial cell line NL-20. CHEMOSPHERE 2024; 362:142622. [PMID: 38880264 DOI: 10.1016/j.chemosphere.2024.142622] [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: 01/07/2024] [Revised: 06/02/2024] [Accepted: 06/14/2024] [Indexed: 06/18/2024]
Abstract
The heterogeneity and complexity of solvent-extracted organic matter associated with PM2.5 (SEOM-PM2.5) is well known; however, there is scarce information on its biological effects in human cells. This work aimed to evaluate the effect of SEOM-PM2.5 collected in northern Mexico City during the cold-dry season (November 2017) on NL-20 cells, a human bronchial epithelial cell line. The SEOM obtained accounted for 15.5% of the PM2.5 mass and contained 21 polycyclic aromatic hydrocarbons (PAHs). The cell viability decreased following exposure to SEOM-PM2.5, and there were noticeable morphological changes such as increased cell size and the presence of cytoplasmic vesicles in cells treated with 5-40 μg/mL SEOM-PM2.5. Exposure to 5 μg/mL SEOM-PM2.5 led to several alterations compared with the control cells, including the induction of double-stranded DNA breaks based (p < 0.001); nuclear fragmentation and an increased mitotic index (p < 0.05); 53BP1 staining, a marker of DNA repair by non-homologous end-joining (p < 0.001); increased BiP protein expression; and reduced ATF6, IRE1α, and PERK gene expression. Conversely, when exposed to 40 μg/mL SEOM-PM2.5, the cells showed an increase in reactive oxygen species formation (p < 0.001), BiP protein expression (p < 0.05), and PERK gene expression (p < 0.05), indicating endoplasmic reticulum stress. Our data suggest concentration-dependent toxicological effects of SEOM-PM2.5 on NL-20 cells, including genotoxicity, genomic instability, and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Josefina Poblano-Bata
- Especiación Química de Aerosoles Orgánicos Atmosféricos, Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México. Investigación Científica s/n, C.U., Coyoacán, Mexico City, 04510, Mexico; Centro de Investigación en Biomedicina y Bioseguridad, Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City, 06720, Mexico.
| | - Montserrat Zaragoza-Ojeda
- Centro de Investigación en Biomedicina y Bioseguridad, Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City, 06720, Mexico.
| | - Andrea De Vizcaya-Ruiz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados-IPN, Ciudad de México, 07360, Mexico.
| | - Francisco Arenas-Huertero
- Centro de Investigación en Biomedicina y Bioseguridad, Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City, 06720, Mexico.
| | - Omar Amador-Muñoz
- Especiación Química de Aerosoles Orgánicos Atmosféricos, Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México. Investigación Científica s/n, C.U., Coyoacán, Mexico City, 04510, Mexico.
| |
Collapse
|
35
|
Falvo S, Grillo G, Latino D, Chieffi Baccari G, Di Fiore MM, Venditti M, Petito G, Santillo A. Potential role of mitochondria and endoplasmic reticulum in the response elicited by D-aspartate in TM4 Sertoli cells. Front Cell Dev Biol 2024; 12:1438231. [PMID: 39105170 PMCID: PMC11298366 DOI: 10.3389/fcell.2024.1438231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/04/2024] [Indexed: 08/07/2024] Open
Abstract
D-Aspartic Acid (D-Asp) affects spermatogenesis by enhancing the biosynthesis of the sex steroid hormones acting either through the hypothalamus-pituitary-testis axis or directly on Leydig cells. Recently, in vitro studies have also demonstrated the direct effects of D-Asp on the proliferation and/or activity of germ cells. However, although D-Asp is present in Sertoli cells (SC), the specific role of the amino acid in these cells remains unknown. This study investigated the effects of D-Asp on the proliferation and activity of TM4 SC, focusing on the mitochondrial compartment and its association with the endoplasmic reticulum (ER). We found that D-Asp enhanced the proliferation and activity of TM4 cells as evidenced by the activation of ERK/Akt/PCNA pathway and the increase in the protein levels of the androgen receptor. Furthermore, D-Asp reduced both the oxidative stress and apoptotic process. An increase in mitochondrial functionality and dynamics, as well as a reduction in ER stress, were also found in D-Asp-treated TM4 cells. It is known that mitochondria are closely associated with ER to form the Mitochondrial-Associated Endoplasmic Reticulum Membranes (MAM), the site of calcium ions and lipid transfer from ER to the mitochondria, and vice versa. The data demonstrated that D-Asp induced stabilization of MAM in TM4 cells. In conclusion, this study is the first to demonstrate a direct effect of D-Asp on SC activity and to clarify the cellular/molecular mechanism underlying these effects, suggesting that D-Asp could stimulate spermatogenesis by improving the efficiency of SC.
Collapse
Affiliation(s)
- Sara Falvo
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Giulia Grillo
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Debora Latino
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Gabriella Chieffi Baccari
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Maria Maddalena Di Fiore
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Massimo Venditti
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuseppe Petito
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Alessandra Santillo
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy
| |
Collapse
|
36
|
Schultheis N, Connell A, Kapral A, Becker RJ, Mueller R, Shah S, O'Donnell M, Roseman M, Swanson L, DeGuara S, Wang W, Yin F, Saini T, Weiss RJ, Selleck SB. Altering heparan sulfate suppresses cell abnormalities and neuron loss in Drosophila presenilin model of Alzheimer Disease. iScience 2024; 27:110256. [PMID: 39109174 PMCID: PMC11302002 DOI: 10.1016/j.isci.2024.110256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/06/2024] [Accepted: 06/10/2024] [Indexed: 08/10/2024] Open
Abstract
We examined the function of heparan-sulfate-modified proteoglycans (HSPGs) in pathways affecting Alzheimer disease (AD)-related cell pathology in human cell lines and mouse astrocytes. Mechanisms of HSPG influences on presenilin-dependent cell loss were evaluated in Drosophila using knockdown of the presenilin homolog, Psn, together with partial loss-of-function of sulfateless (sfl), a gene specifically affecting HS sulfation. HSPG modulation of autophagy, mitochondrial function, and lipid metabolism were shown to be conserved in human cell lines, Drosophila, and mouse astrocytes. RNA interference (RNAi) of Ndst1 reduced intracellular lipid levels in wild-type mouse astrocytes or those expressing humanized variants of APOE, APOE3, and APOE4. Neuron-directed knockdown of Psn in Drosophila produced apoptosis and cell loss in the brain, phenotypes suppressed by reductions in sfl expression. Abnormalities in mitochondria, liposomes, and autophagosome-derived structures in animals with Psn knockdown were also rescued by reduction of sfl. These findings support the direct involvement of HSPGs in AD pathogenesis.
Collapse
Affiliation(s)
- Nicholas Schultheis
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Alyssa Connell
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Alexander Kapral
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Robert J. Becker
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Richard Mueller
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Shalini Shah
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Mackenzie O'Donnell
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Matthew Roseman
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Lindsey Swanson
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Sophia DeGuara
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Weihua Wang
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, AZ 85721, USA
| | - Fei Yin
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, AZ 85721, USA
| | - Tripti Saini
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Ryan J. Weiss
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Scott B. Selleck
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
37
|
Chen J, Chen X, Tang A, Wang Z, Cheong KL, Liu X, Zhong S. Chondroitin sulfate-functionalized selenium nanoparticle-induced S-phase cell cycle arrest and apoptosis in HeLa Cells. J Food Sci 2024; 89:4469-4479. [PMID: 38837700 DOI: 10.1111/1750-3841.17137] [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: 01/31/2024] [Revised: 04/09/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024]
Abstract
This study aimed to evaluate the anti-cervical cancer activity of chondroitin sulfate-functionalized selenium nanoparticles (SeCS) and to elucidate their action mechanism. Cytotoxic effect of SeCS on HeLa cells was assessed by MTT assay. Further molecular mechanism of SeCS was analyzed by flow cytometric assay and western blotting. The results showed that treatment with SeCS resulted in a dose- and time-dependent inhibition in the proliferation of HeLa cells. The data obtained from flow cytometry demonstrated that SeCS inhibited HeLa cell growth via the induction of S-phase arrest and cell apoptosis. Further mechanism analysis found that SeCS down-regulated expression levels of cyclin A and CDK2 and up-regulated p21 expression, which contributed to S arrest. Moreover, SeCS increased the level of Bax and decreased the expression of Bcl-2, resulting in the release of cytochrome C from mitochondria and activating caspase-3/8/9 for caspase-dependent apoptosis. Meanwhile, intracellular reactive oxygen species (ROS) levels were elevated after SeCS treatment, suggesting that ROS might be upstream of SeCS-induced S-phase arrest and cell apoptosis. These data show that SeCS has anti-tumor effects and possesses the potential to become a new therapeutic agent or adjuvant therapy for cancer patients. PRACTICAL APPLICATION: In our previous study, we used chondroitin sulfate to stabilize nano-selenium to obtain SeCS to improve the bioactivity and stability of nano-selenium. We found that it possessed an inhibitory effect on HeLa cells. However, the molecular mechanism remains unclear. This study elucidated the mechanism of SeCS damage to HeLa cells. SeCS has the potential to become a new therapeutic agent or adjuvant therapy for cancer patients.
Collapse
Affiliation(s)
- Jianping Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Xuehua Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Anqi Tang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Zhuo Wang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Kit-Leong Cheong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Xiaofei Liu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Saiyi Zhong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
38
|
Chota A, George BP, Abrahamse H. Apoptotic efficiency of Dicoma anomala biosynthesized silver nanoparticles against A549 lung cancer cells. Biomed Pharmacother 2024; 176:116845. [PMID: 38810403 DOI: 10.1016/j.biopha.2024.116845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024] Open
Abstract
Lung cancer is one of the common forms of cancer that affects both men and women and is regarded as the leading cause of cancer related deaths. It is characterized by unregulated cell division of altered cells within the lung tissues. Green nanotechnology is a promising therapeutic option that is adopted in cancer research. Dicoma anomala (D. anomala) is one of the commonly used African medicinal plant in the treatment of different medical conditions including cancer. In the present study, silver nanoparticles (AgNPs) were synthesized using D. anomala MeOH root extract. We evaluated the anticancer efficacy of the synthesized AgNPs as an individual treatment as well as in combination with pheophorbide a (PPBa) mediated photodynamic therapy (PDT) in vitro. UV-VIS spectroscopy, high-resolution transmission electron microscopy (HR-TEM), Scanning electron microscopy (SEM) and energy dispersive X-ray spectroscopy (EDS) was used to confirm the formation of D.A AgNPs. Post 24 h treatment, A549 cells were evaluated for ATP proliferation, morphological changes supported by LIVE/DEAD assay, and caspase activities. All experiments were repeated four times (n=4), with findings being analysed using SPSS statistical software version 27 set at 0.95 confidence interval. The results from the present study revealed a dose-dependent decrease in cell proliferation in both individual and combination therapy of PPBa mediated PDT and D.A AgNPs on A549 lung cancer cells with significant morphological changes. Additionally, LIVE/DEAD assay displayed a significant increase in the number of dead cell population in individual treatments (i.e., IC50's treated A549 cells) as well as in combination therapy. In conclusion, the findings from this study demonstrated the anticancer efficacy of green synthesized AgNPs as a mono-therapeutic drug as well as in combination with a chlorophyll derivative PPBa in PDT. Taken together, the findings highlight the therapeutic potential of green nanotechnology in medicine.
Collapse
Affiliation(s)
- Alexander Chota
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 1711, Doornfontein, Johannesburg 2028, South Africa
| | - Blassan P George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 1711, Doornfontein, Johannesburg 2028, South Africa.
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 1711, Doornfontein, Johannesburg 2028, South Africa
| |
Collapse
|
39
|
Petersilie L, Heiduschka S, Nelson JS, Neu LA, Le S, Anand R, Kafitz KW, Prigione A, Rose CR. Cortical brain organoid slices (cBOS) for the study of human neural cells in minimal networks. iScience 2024; 27:109415. [PMID: 38523789 PMCID: PMC10957451 DOI: 10.1016/j.isci.2024.109415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/29/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Brain organoids derived from human pluripotent stem cells are a promising tool for studying human neurodevelopment and related disorders. Here, we generated long-term cultures of cortical brain organoid slices (cBOS) grown at the air-liquid interphase from regionalized cortical organoids. We show that cBOS host mature neurons and astrocytes organized in complex architecture. Whole-cell patch-clamp demonstrated subthreshold synaptic inputs and action potential firing of neurons. Spontaneous intracellular calcium signals turned into synchronous large-scale oscillations upon combined disinhibition of NMDA receptors and blocking of GABAA receptors. Brief metabolic inhibition to mimic transient energy restriction in the ischemic brain induced reversible intracellular calcium loading of cBOS. Moreover, metabolic inhibition induced a reversible decline in neuronal ATP as revealed by ATeam1.03YEMK. Overall, cBOS provide a powerful platform to assess morphological and functional aspects of human neural cells in intact minimal networks and to address the pathways that drive cellular damage during brain ischemia.
Collapse
Affiliation(s)
- Laura Petersilie
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Sonja Heiduschka
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital and Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Joel S.E. Nelson
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Louis A. Neu
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Stephanie Le
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital and Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Karl W. Kafitz
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital and Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Christine R. Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| |
Collapse
|
40
|
Lin H, Chung M, Sun J, Yang Y, Zhang L, Pan X, Wei M, Cai S, Pan Y. Ganoderma spore lipid ameliorates docetaxel, cisplatin, and 5-fluorouracil chemotherapy-induced damage to bone marrow mesenchymal stem cells and hematopoiesis. BMC Complement Med Ther 2024; 24:158. [PMID: 38610025 PMCID: PMC11010295 DOI: 10.1186/s12906-024-04445-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND A triplet chemotherapy regimen of docetaxel, cisplatin, and 5-fluorouracil (TPF) is used to treat head and neck squamous cell carcinoma; however, it is toxic to bone marrow mesenchymal stem cells (BMSCs). We previously demonstrated that Ganoderma spore lipid (GSL) protect BMSCs against cyclophosphamide toxicity. In this study, we investigated the protective effects of GSL against TPF-induced BMSCs and hematopoietic damage. METHODS BMSCs and C57BL/6 mice were divided into control, TPF, co-treatment (simultaneously treated with GSL and TPF for 2 days), and pre-treatment (treated with GSL for 7 days before 2 days of TPF treatment) groups. In vitro, morphology, phenotype, proliferation, senescence, apoptosis, reactive oxygen species (ROS), and differentiation of BMSCs were evaluated. In vivo, peripheral platelets (PLTs) and white blood cells (WBCs) from mouse venous blood were quantified. Bone marrow cells were isolated for hematopoietic colony-forming examination. RESULTS In vitro, GSL significantly alleviated TPF-induced damage to BMSCs compared with the TPF group, recovering their morphology, phenotype, proliferation, and differentiation capacity (p < 0.05). Annexin V/PI and senescence-associated β-galactosidase staining showed that GSL inhibited apoptosis and delayed senescence in TPF-treated BMSCs (p < 0.05). GSL downregulated the expression of caspase-3 and reduced ROS formation (p < 0.05). In vivo, GSL restored the number of peripheral PLTs and WBCs and protected the colony-forming capacity of bone marrow cells (p < 0.05). CONCLUSIONS GSL efficiently protected BMSCs from damage caused by TPF and recovered hematopoiesis.
Collapse
Affiliation(s)
- Haohui Lin
- Health Science Center, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Manhon Chung
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingchun Sun
- Department of Head and Neck Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yi Yang
- Health Science Center, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Li Zhang
- Health Science Center, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Xiaohua Pan
- Health Science Center, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Minghui Wei
- Department of Head and Neck Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.
| | - Sa Cai
- Health Science Center, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China.
| | - Yu Pan
- Health Science Center, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China.
| |
Collapse
|
41
|
Abe J, Aono Y, Hirai Y. The decline in cellular iron is crucial for differentiation in keratinocytes. Metallomics 2024; 16:mfae014. [PMID: 38449344 DOI: 10.1093/mtomcs/mfae014] [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] [Accepted: 03/05/2024] [Indexed: 03/08/2024]
Abstract
Iron is a vital metal for most biological functions in tissues, and its concentration is exquisitely regulated at the cellular level. During the process of differentiation, keratinocytes in the epidermis undergo a noticeable reduction in iron content. Conversely, psoriatic lesions, characterized by disruptions in epidermal differentiation, frequently reveal an excessive accumulation of iron within keratinocytes that have undergone differentiation. In this study, we clarified the significance of attenuated cellular iron content in the intricate course of epidermal differentiation. We illustrated this phenomenon through the utilization of hinokitiol, an iron chelator derived from the heartwood of Taiwanese hinoki, which forcibly delivers iron into cells independent of the intrinsic iron-regulation systems. While primary cultured keratinocytes readily succumbed to necrotic cell death by this iron chelator, mild administration of the hinokitiol-iron complex modestly disrupts the process of differentiation in these cells. Notably, keratinocyte model cells HaCaT and anaplastic skin rudiments exhibit remarkable resilience against the cytotoxic impact of hinokitiol, and the potent artificial influx of iron explains a suppressive effect selectively on epidermal differentiation. Moreover, the augmentation of iron content induced by the overexpression of divalent metal transporter 1 culminates in the inhibition of differentiation in HaCaT cells. Consequently, the diminution in cellular iron content emerges as an important determinant influencing the trajectory of keratinocyte differentiation.
Collapse
Affiliation(s)
- Junya Abe
- Department of Biomedical Sciences, Graduate School of Science and Technology, Kwansei Gakuin University. 1, Gakuen-Uegahara, Sanda 669-1330, Japan
| | - Yuichi Aono
- Department of Biomedical Sciences, Graduate School of Science and Technology, Kwansei Gakuin University. 1, Gakuen-Uegahara, Sanda 669-1330, Japan
| | - Yohei Hirai
- Department of Biomedical Sciences, Graduate School of Science and Technology, Kwansei Gakuin University. 1, Gakuen-Uegahara, Sanda 669-1330, Japan
| |
Collapse
|
42
|
Ma J, Chen H. Efficient Supervised Pretraining of Swin-Transformer for Virtual Staining of Microscopy Images. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:1388-1399. [PMID: 38010933 DOI: 10.1109/tmi.2023.3337253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Fluorescence staining is an important technique in life science for labeling cellular constituents. However, it also suffers from being time-consuming, having difficulty in simultaneous labeling, etc. Thus, virtual staining, which does not rely on chemical labeling, has been introduced. Recently, deep learning models such as transformers have been applied to virtual staining tasks. However, their performance relies on large-scale pretraining, hindering their development in the field. To reduce the reliance on large amounts of computation and data, we construct a Swin-transformer model and propose an efficient supervised pretraining method based on the masked autoencoder (MAE). Specifically, we adopt downsampling and grid sampling to mask 75% of pixels and reduce the number of tokens. The pretraining time of our method is only 1/16 compared with the original MAE. We also design a supervised proxy task to predict stained images with multiple styles instead of masked pixels. Additionally, most virtual staining approaches are based on private datasets and evaluated by different metrics, making a fair comparison difficult. Therefore, we develop a standard benchmark based on three public datasets and build a baseline for the convenience of future researchers. We conduct extensive experiments on three benchmark datasets, and the experimental results show the proposed method achieves the best performance both quantitatively and qualitatively. In addition, ablation studies are conducted, and experimental results illustrate the effectiveness of the proposed pretraining method. The benchmark and code are available at https://github.com/birkhoffkiki/CAS-Transformer.
Collapse
|
43
|
Thepthanee C, Ei ZZ, Benjakul S, Zou H, Petsri K, Innets B, Chanvorachote P. Shrimp Lipids Inhibit Migration, Epithelial-Mesenchymal Transition, and Cancer Stem Cells via Akt/mTOR/c-Myc Pathway Suppression. Biomedicines 2024; 12:722. [PMID: 38672078 PMCID: PMC11048134 DOI: 10.3390/biomedicines12040722] [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: 02/09/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Shrimp is a rich source of bioactive molecules that provide health benefits. However, the high cholesterol content in shrimp oil may pose a risk. We utilized the cholesterol elimination method to obtain cholesterol-free shrimp lipids (CLs) and investigated their anticancer potential, focusing on cancer stem cells (CSCs) and epithelial-to-mesenchymal transition (EMT). Our study focused on CSCs and EMT, as these factors are known to contribute to cancer metastasis. The results showed that treatment with CLs at doses ranging from 0 to 500 µg/mL significantly suppressed the cell migration ability of human lung cancer (H460 and H292) cells, indicating its potential to inhibit cancer metastasis. The CLs at such concentrations did not cause cytotoxicity to normal human keratinocytes. Additionally, CL treatment was found to significantly reduce the levels of Snail, Slug, and Vimentin, which are markers of EMT. Furthermore, we investigated the effect of CLs on CSC-like phenotypes and found that CLs could significantly suppress the formation of a three-dimensional (3D) tumor spheroid in lung cancer cells. Furthermore, CLs induced apoptosis in the CSC-rich population and significantly depleted the levels of CSC markers CD133, CD44, and Sox2. A mechanistic investigation demonstrated that exposing lung cancer cells to CLs downregulated the phosphorylation of Akt and mTOR, as well as c-Myc expression. Based on these findings, we believe that CLs may have beneficial effects on health as they potentially suppress EMT and CSCs, as well as the cancer-potentiating pathway of Akt/mTOR/c-Myc.
Collapse
Affiliation(s)
- Chorpaka Thepthanee
- Department of Food Science, School of Food Industry, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| | - Zin Zin Ei
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (Z.Z.E.); (B.I.)
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkhla University, Songkhla 90110, Thailand;
| | - Hongbin Zou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China;
| | - Korrakod Petsri
- Department of Pharmacology, Faculty of Medicine, Kasetsart University, Bangkok 10900, Thailand;
| | - Bhurichaya Innets
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (Z.Z.E.); (B.I.)
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pithi Chanvorachote
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (Z.Z.E.); (B.I.)
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
44
|
Pulfer A, Pizzagalli DU, Gagliardi PA, Hinderling L, Lopez P, Zayats R, Carrillo-Barberà P, Antonello P, Palomino-Segura M, Grädel B, Nicolai M, Giusti A, Thelen M, Gambardella LM, Murooka TT, Pertz O, Krause R, Gonzalez SF. Transformer-based spatial-temporal detection of apoptotic cell death in live-cell imaging. eLife 2024; 12:RP90502. [PMID: 38497754 PMCID: PMC10948145 DOI: 10.7554/elife.90502] [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] [Indexed: 03/19/2024] Open
Abstract
Intravital microscopy has revolutionized live-cell imaging by allowing the study of spatial-temporal cell dynamics in living animals. However, the complexity of the data generated by this technology has limited the development of effective computational tools to identify and quantify cell processes. Amongst them, apoptosis is a crucial form of regulated cell death involved in tissue homeostasis and host defense. Live-cell imaging enabled the study of apoptosis at the cellular level, enhancing our understanding of its spatial-temporal regulation. However, at present, no computational method can deliver robust detection of apoptosis in microscopy timelapses. To overcome this limitation, we developed ADeS, a deep learning-based apoptosis detection system that employs the principle of activity recognition. We trained ADeS on extensive datasets containing more than 10,000 apoptotic instances collected both in vitro and in vivo, achieving a classification accuracy above 98% and outperforming state-of-the-art solutions. ADeS is the first method capable of detecting the location and duration of multiple apoptotic events in full microscopy timelapses, surpassing human performance in the same task. We demonstrated the effectiveness and robustness of ADeS across various imaging modalities, cell types, and staining techniques. Finally, we employed ADeS to quantify cell survival in vitro and tissue damage in mice, demonstrating its potential application in toxicity assays, treatment evaluation, and inflammatory dynamics. Our findings suggest that ADeS is a valuable tool for the accurate detection and quantification of apoptosis in live-cell imaging and, in particular, intravital microscopy data, providing insights into the complex spatial-temporal regulation of this process.
Collapse
Affiliation(s)
- Alain Pulfer
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USILuganoSwitzerland
- Department of Information Technology and Electrical Engineering, ETH ZurichZürichSwitzerland
| | - Diego Ulisse Pizzagalli
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USILuganoSwitzerland
- Euler Institute, USILuganoSwitzerland
| | | | | | | | | | - Pau Carrillo-Barberà
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USILuganoSwitzerland
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universitat de ValènciaValenciaSpain
| | - Paola Antonello
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USILuganoSwitzerland
- Institute of Cell Biology, University of BernBernSwitzerland
| | | | - Benjamin Grädel
- Institute of Cell Biology, University of BernBernSwitzerland
| | | | - Alessandro Giusti
- Dalle Molle Institute for Artificial Intelligence, IDSIALuganoSwitzerland
| | - Marcus Thelen
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USILuganoSwitzerland
| | | | | | - Olivier Pertz
- Institute of Cell Biology, University of BernBernSwitzerland
| | | | | |
Collapse
|
45
|
Yoon J, Kim BS. Coordinative Double Hydrophilic All-Polyether Micelles for pH-Responsive Delivery of Cisplatin. Biomacromolecules 2024; 25:1861-1870. [PMID: 38344950 DOI: 10.1021/acs.biomac.3c01301] [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: 03/12/2024]
Abstract
Despite its widespread use in the treatment of numerous cancers, the use of cisplatin still raises concerns about its high toxicity and limited selectivity. Consequently, the necessity arises for the development of an effective drug delivery system. Here, we present an effective approach that introduces a double hydrophilic block copolyether for the controlled delivery of cisplatin. Specifically, poly(ethylene glycol)-block-poly(glycidoxy acetic acid) (mPEG-b-PGA) was synthesized via anionic ring-opening polymerization using the oxazoline-based epoxide monomer 4,4-dimethyl-2-oxazoline glycidyl ether, followed by subsequent acidic deprotection. The coordinative metal-ligand interaction between cisplatin and the carboxylate group within the PGA block facilitated the formation of micelles from the double hydrophilic mPEG-b-PGA copolyether. Cisplatin-loaded polymeric micelles had a high loading capacity, controlled pH-responsive release kinetics, and high cell viability. Furthermore, in vitro biological assays revealed cellular apoptosis induced by the cisplatin-loaded micelles. This study thus successfully demonstrates the potential use of double hydrophilic block copolyethers as a versatile platform for biomedical applications.
Collapse
Affiliation(s)
- Jiwoo Yoon
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Byeong-Su Kim
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
46
|
Hussain Y, Singh J, Meena A, Sinha RA, Luqman S. Escin-sorafenib synergy up-regulates LC3-II and p62 to induce apoptosis in hepatocellular carcinoma cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:840-856. [PMID: 37853854 DOI: 10.1002/tox.23988] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a common solid cancer and the leading cause of cancer deaths worldwide. Sorafenib is the first drug used to treat HCC but its effectiveness needs to be improved, and it is important to find ways to treat cancer that combine sorafenib with other drugs. Synergistic therapies lower effective drug doses and side effects while enhancing the anticancer effect. PURPOSE In the present study, the therapeutic potential of sorafenib in combination with escin and its underlying mechanism in targeting liver cancer has been established. STUDY DESIGN/METHODS The IC50 of sorafenib and escin against HepG2, PLC/PRF5 and Huh7 cell lines were determined using MTT assay. The combination index, dose reduction index, isobologram and concentrations producing synergy were evaluated using the Chou-Talaly algorithm. The sub-effective concentration of sorafenib and escin was selected to analyze cytotoxic synergistic potential. Cellular ROS, mitochondrial membrane potential, annexin V and cell cycle were evaluated using a flow-cytometer, and autophagy biomarkers were determined using western blotting. Moreover, autophagy was knocked down using ATG5 siRNA to confirm its role. A DEN-induced liver cancer rat model was developed to check the synergy of sorafenib and escin. RESULTS Different concentrations of escin reduced the IC50 of sorafenib in HepG2, PLC/PRF5 and Huh7 cell lines. Chou-Talaly algorithm determined cytotoxic synergistic concentrations of sorafenib and escin in these cell lines. Mechanistically, this combination over-expressed p62 and LC-II, reflecting autophagy block and induced late apoptosis, further reconfirmed by ATG5 knockdown. Sorafenib and escin combination reduced HCC serum biomarker α-feto protein (α-FP) by 1.5 folds. This combination restricted liver weight, tumor number and size, also, conserved morphological features of liver cells. The combination selectively targeted the G0 /G1 phase of cancer cells. CONCLUSION Escin and sorafenib combination potentially up-regulates p62 to block autophagy to induce late apoptosis in liver cancer cells.
Collapse
Affiliation(s)
- Yusuf Hussain
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Jyoti Singh
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
47
|
Huang M, Chow CH, Gurdita A, Harada H, Pham Truong VQB, Eide S, Sun HS, Feng ZP, Monnier PP, Wallace VA, Sugita S. SNAP-25, but not SNAP-23, is essential for photoreceptor development, survival, and function in mice. Commun Biol 2024; 7:34. [PMID: 38182732 PMCID: PMC10770054 DOI: 10.1038/s42003-023-05760-8] [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: 09/18/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
SNARE-mediated vesicular transport is thought to play roles in photoreceptor glutamate exocytosis and photopigment delivery. However, the functions of Synaptosomal-associated protein (SNAP) isoforms in photoreceptors are unknown. Here, we revisit the expression of SNAP-23 and SNAP-25 and generate photoreceptor-specific knockout mice to investigate their roles. Although we find that SNAP-23 shows weak mRNA expression in photoreceptors, SNAP-23 removal does not affect retinal morphology or vision. SNAP-25 mRNA is developmentally regulated and undergoes mRNA trafficking to photoreceptor inner segments at postnatal day 9 (P9). SNAP-25 knockout photoreceptors develop normally until P9 but degenerate by P14 resulting in severe retinal thinning. Photoreceptor loss in SNAP-25 knockout mice is associated with abolished electroretinograms and vision loss. We find mistrafficked photopigments, enlarged synaptic vesicles, and abnormal synaptic ribbons which potentially underlie photoreceptor degeneration. Our results conclude that SNAP-25, but not SNAP-23, mediates photopigment delivery and synaptic functioning required for photoreceptor development, survival, and function.
Collapse
Affiliation(s)
- Mengjia Huang
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Chun Hin Chow
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Akshay Gurdita
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Hidekiyo Harada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Victor Q B Pham Truong
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Sarah Eide
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Anatomy, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Philippe P Monnier
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Department of Ophthalmology & Vision Sciences, University of Toronto, Toronto, ON, M5T 3A9, Canada
| | - Valerie A Wallace
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Department of Ophthalmology & Vision Sciences, University of Toronto, Toronto, ON, M5T 3A9, Canada
| | - Shuzo Sugita
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON, M5T 0S8, Canada.
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
48
|
Bahadır Semerci A, Yıldırım M, Oztay F, Sagıroglu M, Tunç K. Evaluation of Fatty Acid Contents and Biological Activities of Jurinea turcica. Chem Biodivers 2024; 21:e202300084. [PMID: 38010957 DOI: 10.1002/cbdv.202300084] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 11/29/2023]
Abstract
The fatty acid profile, antioxidant/antibacterial, and cytotoxic effects of the extracts obtained from Jurinea turcica B.Doğan& A.Duran have been evaluated for the first time in the current study. The fatty acid profile of ethanolic extracts was determined using the Soxhlet extractor by a gas chromatography-mass spectrometer. The antioxidant and antibacterial activities were measured by 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging and ferrous reduction tests and the disc diffusion technique. Additionally, the cytotoxicity and wound healing assays were performed on A549 cells. The highest amount of component in the leaf extract was docosanoic acid methyl ester, whereas abundant arachidonic acid methyl ester was mainly found in the flower extract. The IC50 values, the 50 % scavenging value for the DPPH radical, were 179.13 and 124.67 μg/mL for the leaf and flower extracts, respectively. IC50 values (the half-maximal inhibitory concentration) were 10.4 and 24.7 μg/mL for the flower and leaf extracts, respectively. The leaf extract showed more potent antibacterial activity on Enterococcus faecalis (17 mm) and Staphylococcus aureus (16 mm) bacteria than the flower extract. In conclusion, the extracts of J. turcica have anti-cancerogenic and antibacterial effects. Leaf extracts have antibacterial and anti-metastatic effects, while flower extracts show antioxidant, cytotoxic, and apoptotic properties in A549 cells.
Collapse
Affiliation(s)
- Alican Bahadır Semerci
- Sakarya University, Science Faculty, Department of Biology, 54187, Sakarya, Turkey
- Necmettin Erbakan University, Ereğli Vocational School of Health Services, 42310, Konya, Turkey
| | - Merve Yıldırım
- Istanbul University, Science Faculty, Department of Biology, Molecular Biology Division, Vezneciler, 34134, Istanbul, Turkey
| | - Füsun Oztay
- Istanbul University, Science Faculty, Department of Biology, Molecular Biology Division, Vezneciler, 34134, Istanbul, Turkey
| | - Mehmet Sagıroglu
- Sakarya University, Science Faculty, Department of Biology, 54187, Sakarya, Turkey
| | - Kenan Tunç
- Sakarya University, Science Faculty, Department of Biology, 54187, Sakarya, Turkey
| |
Collapse
|
49
|
Singhal S, Rani V. Cardioprotective Role of Tinospora cordifolia against Trimethylamine-N-Oxide and Glucose Induced Stress in Rat Cardiomyocytes. Cardiovasc Hematol Agents Med Chem 2024; 22:475-494. [PMID: 37907489 DOI: 10.2174/0118715257270512231013064533] [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: 07/04/2023] [Revised: 08/22/2023] [Accepted: 09/15/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND Type 2 diabetes has become a concern issue that affects the quality of life and can increase the risk of cardiac insufficiency elevating the threat to the life safety of patients. A recognized cause of cardiac insufficiency is diabetic cardiomyopathy, chronic hyperglycemia, and myocardial lipotoxicity which can reduce the myocardial contractile performance, and enhance the cardiomyocyte hypertrophy and interstitial fibrosis. The cause of diabetic cardiomyopathy is multi-factorial which includes oxidative stress, insulin resistance, inflammation, apoptosis, and autophagy. Recent clinical studies have suggested the dysbiosis of gut microbiota, secretion of metabolites, and their diffusion in to the host as to have direct detrimental effects on the cardiac contractility. MATERIALS AND METHODS In the present paper, we have done in silico studies including molecular interaction of phytoconstituents of Tinospora cordifolia against reactive oxygen species producing proteins. Whereas, in vitro studies were conducted on H9C2 cardiac cells including cell morphological examination, level of reactive oxygen species, cell count-viability, apoptotic status, in the presence of high glucose, trimethylamine-n-oxide, and plant extracts which were determined through cell analyzer and microscopic assays. RESULTS The treatment of high glucose and trimethylamine-n-oxide was found to be increase the cardiac stress approximately two fold by attenuating hypertrophic conditions, oxidative stress, and apoptosis in rat cardiomyocytes, and Tinospora cordifolia was found to be a cardioprotective agent. CONCLUSION Conclusively, our study has reported that the Indian medicinal plant Tinospora cordifolia has the ability to treat diabetic cardiomyopathy. Our study can open up a new herbal therapeutic strategy against diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Shivani Singhal
- Center for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector- 62, Noida, 201307, Uttar Pradesh, India
| | - Vibha Rani
- Center for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector- 62, Noida, 201307, Uttar Pradesh, India
| |
Collapse
|
50
|
Jung EJ, Kim HJ, Shin SC, Kim GS, Jung JM, Hong SC, Kim CW, Lee WS. Artemisia annua L. Polyphenols Enhance the Anticancer Effect of β-Lapachone in Oxaliplatin-Resistant HCT116 Colorectal Cancer Cells. Int J Mol Sci 2023; 24:17505. [PMID: 38139333 PMCID: PMC10743427 DOI: 10.3390/ijms242417505] [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: 10/31/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Recent studies suggest that the anticancer activity of β-lapachone (β-Lap) could be improved by different types of bioactive phytochemicals. The aim of this study was to elucidate how the anticancer effect of β-Lap is regulated by polyphenols extracted from Korean Artemisia annua L. (pKAL) in parental HCT116 and oxaliplatin-resistant (OxPt-R) HCT116 colorectal cancer cells. Here, we show that the anticancer effect of β-Lap is more enhanced by pKAL in HCT116-OxPt-R cells than in HCT116 cells via a CCK-8 assay, Western blot, and phase-contrast microscopy analysis of hematoxylin-stained cells. This phenomenon was associated with the suppression of OxPt-R-related upregulated proteins including p53 and β-catenin, the downregulation of cell survival proteins including TERT, CD44, and EGFR, and the upregulation of cleaved HSP90, γ-H2AX, and LC3B-I/II. A bioinformatics analysis of 21 proteins regulated by combined treatment of pKAL and β-Lap in HCT116-OxPt-R cells showed that the enhanced anticancer effect of β-Lap by pKAL was related to the inhibition of negative regulation of apoptotic process and the induction of DNA damage through TERT, CD44, and EGFR-mediated multiple signaling networks. Our results suggest that the combination of pKAL and β-Lap could be used as a new therapy with low toxicity to overcome the OxPt-R that occurred in various OxPt-containing cancer treatments.
Collapse
Affiliation(s)
- Eun Joo Jung
- Department of Internal Medicine, Institute of Medical Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, 15 Jinju-daero 816 Beon-gil, Jinju 52727, Republic of Korea;
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Medical Science, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
| | - Sung Chul Shin
- Department of Chemistry, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Gon Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Jin-Myung Jung
- Department of Neurosurgery, Institute of Medical Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea;
| | - Soon Chan Hong
- Department of Surgery, Institute of Medical Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea;
| | - Choong Won Kim
- Department of Biochemistry, Institute of Medical Science, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea;
| | - Won Sup Lee
- Department of Internal Medicine, Institute of Medical Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, 15 Jinju-daero 816 Beon-gil, Jinju 52727, Republic of Korea;
| |
Collapse
|