1
|
Sarwareddy KK, Singh AD, Patnam S, Sesuraj BA, Ponamgi S, Thakur BK, Manda VS. Harnessing tomato-derived small extracellular vesicles as drug delivery system for cancer therapy. Future Sci OA 2025; 11:2461956. [PMID: 39920889 PMCID: PMC11812386 DOI: 10.1080/20565623.2025.2461956] [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: 10/05/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
AIM This study aims to explore a sustainable and scalable approach using tomato fruit-derived sEVs (TsEVs) to deliver calcitriol for enhanced anticancer effects, addressing challenges of low yield and high costs associated with mammalian cell-derived sEVs. METHODS TsEVs were isolated by centrifugation and ultrafiltration and characterized using DLS, TEM, and biochemical assays. Calcitriol was loaded into TsEVs via loading methods, with efficiency measured by spectrophotometry and HPLC. HCT116 and HT29 colon cancer cells were treated with TsEV-calcitriol and assessed for viability, colony formation, migration, ROS levels, and apoptosis gene expression. RESULTS Isolated TsEVs ranged from 30-200 nm with a protein-to-lipid ratio of ∼1. Calcitriol encapsulation efficiencies were 15.4% (passive), 34.8% (freeze-thaw), and 47.3% (sonication). TsEV-calcitriol reduced HCT116 cell viability with IC50 values of 4.05 µg/ml (24 h) and 2.07 µg/ml (48 h). Clonogenic assays showed reduced colony formation and migration. Elevated ROS levels and increased Bax/Bcl-2 ratio were observed in treated HCT116 and HT29 colon cancer cells. CONCLUSION These findings highlight TsEVs as a promising alternative drug delivery platform to mammalian cell-derived sEV for enhancing the therapeutic efficiency of calcitriol and other anticancer agents.
Collapse
Affiliation(s)
- Kartik Kumar Sarwareddy
- Apollo Hospitals Educational and Research Foundation (AHERF), Apollo Hospitals, Hyderabad, India
| | - Anula Divyash Singh
- Apollo Hospitals Educational and Research Foundation (AHERF), Apollo Hospitals, Hyderabad, India
| | - Sreekanth Patnam
- Apollo Hospitals Educational and Research Foundation (AHERF), Apollo Hospitals, Hyderabad, India
| | | | - spd Ponamgi
- Department of Biotechnology, AU College of Science and Technology, Andhra University, India
| | | | - Venkata Sasidhar Manda
- Apollo Hospitals Educational and Research Foundation (AHERF), Apollo Hospitals, Hyderabad, India
- Urvogelbio Private Limited, AHERF, Hyderabad, India
| |
Collapse
|
2
|
Zuo CJ, Tian J. Advancing the understanding of the role of apoptosis in lung cancer immunotherapy: Global research trends, key themes, and emerging frontiers. Hum Vaccin Immunother 2025; 21:2488074. [PMID: 40186454 PMCID: PMC11980473 DOI: 10.1080/21645515.2025.2488074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/12/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025] Open
Abstract
Apoptosis is vital for improving the efficacy of lung cancer (LC) immunotherapy by targeting cancer cell elimination. Despite its importance, there is a lack of comprehensive bibliometric studies analyzing global research on apoptosis in LC immunotherapy. This analysis aims to address this gap by highlighting key trends, contributors, and future directions. A total of 969 publications from 1996 to 2024 were extracted from the Web of Science Core Collection. Analysis was conducted using VOSviewer, CiteSpace, and the R package 'bibliometrix.' The study included contributions from 6,894 researchers across 1,469 institutions in 61 countries, with research published in 356 journals. The volume of publications has steadily increased, led by China and the United States, with Sichuan University as the top contributor. The journal Cancers published the most articles, while Cancer Research had the highest co-citations. Yu-Quan Wei was the leading author, and Jemal, A. was the most frequently co-cited. Key research themes include "cell death mechanisms," "immune regulation," "combination therapies," "gene and nanomedicine applications," and "traditional Chinese medicine (TCM)." Future research is likely to focus on "coordinated regulation of multiple cell death pathways," "modulation of the tumor immune microenvironment," "optimization of combination therapies," "novel strategies in gene regulation," and the "integration of TCM" for personalized treatment. This is the first bibliometric analysis on the role of apoptosis in LC immunotherapy, providing an landscape of global research patterns and emerging therapeutic strategies. The findings offer insights to guide future research and optimize treatment approaches.
Collapse
Affiliation(s)
- Chun-Jian Zuo
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Kim JH, Shim J, Ko N, Kim HJ, Lee Y, Bang S, Han A, Lee S, Kim H, Cho J, Choi K. Effect of porcine oviductal fluid-derived extracellular vesicle supplementation on in vitro embryonic developmental competence and the production efficiency of cloned pigs. Theriogenology 2025; 242:117442. [PMID: 40250251 DOI: 10.1016/j.theriogenology.2025.117442] [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: 12/26/2024] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/20/2025]
Abstract
Extracellular vesicles (EVs) derived from oviductal epithelial cells help with fertilization and embryo development. Although the effects of EVs on the developmental competence of somatic cell nuclear transfer (SCNT) embryos are known, their impact on the production efficiency of cloned pigs in surrogate mothers is unclear. We evaluated the effects of porcine oviductal fluid-derived EVs (oEVs) on the in vitro development of SCNT embryos and production efficiency in cloned pigs. Embryos were treated with isolated and characterized oEVs for 48 h, corresponding to the residence time of fertilized embryos in the oviduct. In both parthenogenetic activation (PA) and SCNT embryos, oEV-treated groups exhibited higher rate of blastocyst formation than the controls. Gene expression analysis revealed increased expression of genes essential for early embryonic development in four-cell stage SCNT embryos, including those involved in pluripotency (POU5F1; POU class 5 homeobox 1), genome stability (Zinc finger and SCAN domain containing 4), and DNA damage response (Checkpoint kinase 1). Additionally, gene expression analysis of blastocysts from SCNT embryos showed increased expression of pluripotency (POU5F1) and Wnt signal transduction (Axisinhibitionprotein2). Transfer of day-2 in vitro cultured SCNT embryos into surrogate mothers revealed no significant difference in production efficiencies between the control and experimental groups. Thus, enhanced early embryonic developmental competence observed in vitro following oEV treatment of PA or SCNT embryos did not translate into improved production efficiency of cloned pigs following embryo transfer to surrogate mothers. Hence, the impact of oEV treatment on live birth outcomes requires further investigation.
Collapse
Affiliation(s)
- Jun-Hyeong Kim
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea; Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Joohyun Shim
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea
| | - Nayoung Ko
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea
| | - Hyoung-Joo Kim
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea
| | - Yongjin Lee
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea
| | - Seonggyu Bang
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea; College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ayeong Han
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea; College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sanghoon Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Hyunil Kim
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea
| | - Jongki Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Kimyung Choi
- Optipharm Inc, 63, Osongsaengmyeong 6-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28158, Republic of Korea.
| |
Collapse
|
4
|
Das U, Chanda T, Kumar J, Peter A. Discovery of natural MCL1 inhibitors using pharmacophore modelling, QSAR, docking, ADMET, molecular dynamics, and DFT analysis. Comput Biol Chem 2025; 117:108427. [PMID: 40120151 DOI: 10.1016/j.compbiolchem.2025.108427] [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: 01/30/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
Mcl-1, a member of the Bcl-2 family, is a crucial regulator of apoptosis, frequently overexpressed in various cancers, including lung, breast, pancreatic, cervical, ovarian cancers, leukemia, and lymphoma. Its anti-apoptotic function allows tumor cells to evade cell death and contributes to drug resistance, making it an essential target for anticancer drug development. This study aimed to discover potent antileukemic compounds targeting Mcl-1. We selected diverse molecules from the BindingDB database to construct a structure-based pharmacophore model, which facilitated the virtual screening of 407,270 compounds from the COCONUT database. An e-pharmacophore model was developed using the co-crystallized inhibitor, followed by QSAR modeling to estimate IC50 values and filter compounds with predicted values below the median. The top hits underwent molecular docking and MMGBSA binding energy calculations against Mcl-1, resulting in the selection of two promising candidates for further ADMET analysis. DFT calculations assessed their electronic properties, confirming favorable reactivity profiles of the screened compounds. Predictions for physicochemical and ADMET properties aligned with expected bioactivity and safety. Molecular dynamics simulations further validated their strong binding affinity and stability, positioning them as potential Mcl-1 inhibitors. Our comprehensive computational approach highlights these compounds as promising antileukemic agents, with future in vivo and in vitro validation recommended for further confirmation.
Collapse
Affiliation(s)
- Uddalak Das
- Department of Plant Biotechnology, University of Agricultural Sciences, Bangalore, Bengaluru, Karnataka 560065, India; School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Tathagata Chanda
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Jitendra Kumar
- Biotechnology Industry Research Assistance Council (BIRAC), Department of Biotechnology (DBT), Ministry of Science and Technology, Government of India, Lodhi Road, New Delhi 110020, India
| | - Anitha Peter
- Department of Plant Biotechnology, University of Agricultural Sciences, Bangalore, Bengaluru, Karnataka 560065, India
| |
Collapse
|
5
|
Yadollahi N, Mohamadian T, Esmaeili D, Forohi F. Design of recombinant bacteriocin fusion protein and evaluation of its anticancer and antibacterial activity. Microb Pathog 2025; 205:107633. [PMID: 40311945 DOI: 10.1016/j.micpath.2025.107633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND AND OBJECTIVES Gastrointestinal cancer is the fifth most common cause of cancer in the world, and its treatment is currently challenging. Bacteriocins are cationic proteins that are produced by the ribosome. Many bacteriocins exhibit cytotoxicity against cancer cells and are attractive candidates for future research. In this study, the anticancer and antibacterial activity of a recombinant six bacteriocin fusion protein bifidocin, enterocin, nisin, thermophilin, epidermycin, and colicin (BENTEC) was investigated against the AGS cancer cell line and important gram-positive and gram-negative pathogens. METHODS The NCBI database provided the gene sequences. The specific ligand of the AGS cell line and the anticancer sequences were added to the recombinant construct. The physical-chemical characteristics of recombinant bacteriocin were assessed by bioinformatics methods. Expression of the recombinant vector pET22b was performed in E. coli BL21(DE3). MTT test was used to evaluate the cytotoxicity in AGS cell lines and normal GES-1 cells. Flow cytometry and Real-Time PCR methods were used to investigate the expression changes of apoptotic genes against the AGS cell line. The anti-bacterial activity of BENTC fusion bacteriocin against Escherichia coli and Enterococcus faecalis was evaluated by MIC, MBC, and disc diffusion methods. RESULTS The recombinant BENTEC fusion bacteriocin contains 424 amino acids, a molecular weight of 47KD, and an isoelectric point of 11.17. The GRAVY score is -0.596, indicating the hydrophilic properties of the fusion bacteriocin. MTT results demonstrated that 52 % of AGS cell lines treated with a concentration of 8 μg/mL of BENTEC fusion bacteriocin were destroyed during 24 hours. Flow cytometry showed that recombinant bacteriocin induced 76.4 % of apoptosis in the treated cell lines (63.7 % early apoptosis and 12.7 % late apoptosis). Real-time RT- PCR indicated that recombinant BENTEC increased expression of the Bax and Caspase 3 genes by 4.53 fold and 5.856 fold, respectively, and decreased gene expression of bcl-2 by 0.18 fold in the AGS line cell. The results demonstrated that the MIC and MBC for E. coli and E. faecalis were 5 μg/ml,10 μg/ml,10 μg/ml, and 20 μg/ml, respectively. Disc diffusion showed that the diameters of the growth inhibition zones for E. coli and E. faecalis were 32 mm and 23 mm, respectively. CONCLUSION The results of this study showed that the fusion protein of six bacteriocins linked to the anticancer and the specific ligand sequences was able to induce apoptosis in AGS cell line and have antibacterial activity against important gram-positive and gram-negative pathogens at lower concentrations and with greater specificity. Collectively, BENTEC fusion bacteriocin may be regarded as a promising option for anticancer and antibacterial therapy or prophylaxis.
Collapse
Affiliation(s)
- Narges Yadollahi
- Department of Microbiology, Shahr-e-Quds Branch, Islamic Azad University, Tehran, Iran
| | - Taher Mohamadian
- Department of Microbiology, Shahr-e-Quds Branch, Islamic Azad University, Tehran, Iran
| | - Davoud Esmaeili
- Department of Microbiology and Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Forohi
- Department of Microbiology, Shahr-e-Quds Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
6
|
Sharma L, Nambiar SS, Mohanty A, Patra A, Kumari S, Saini GK. Elucidating the anticancer properties of posaconazole in triple-negative breast cancer through in-silico and in-vitro analysis. Biochem Biophys Res Commun 2025; 771:152001. [PMID: 40403682 DOI: 10.1016/j.bbrc.2025.152001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/13/2025] [Accepted: 05/10/2025] [Indexed: 05/24/2025]
Abstract
The present study focused on repurposing antifungal drugs for cancer treatment. Repurposing refers to the evaluation of an existing drug that has been used to treat a specific disease and to identify its potential to be used for other therapeutic purposes due to similarities in targets. Posaconazole is a second-generation lipophilic antifungal agent that exhibits broad-spectrum activity against various fungal infections associated with several fungal species. Posaconazole was chosen as the target drug for proteins screened through Swiss Pred to inhibit triple-negative breast cancer. The selected targets were allowed to develop a PPI network using a string database to identify bottleneck proteins. These target proteins were used for molecular docking and molecular dynamics simulations. In-silico analysis was further validated by cell viability assay in which posaconazole exhibited notable decrease in cell viability with an IC50 value of 7.2 μM in MDA-MB-231 triple negative breast cancer cell lines (TNBC). Furthermore, the migration potential of MDA-MB-231 cells following posaconazole treatment was investigated using a wound healing assay. Additionally, Posaconazole caused a notable decrease in sphere and colony formation ability. The mechanism of cell death was further analyzed using intracellular reactive oxygen species generation, mitochondrial depolarization, and cell cycle analysis. The results demonstrated that all tested variables including cell viability, migration colony formation, cell cycle analysis, ROS generation, mitochondrial depolarization and apoptosis showed statistically significant changes (p < 0.01) against the untreated control groups. The potent anticancer potential of posaconazole has opened new avenues for repurposing antifungal drugs to treat cancer.
Collapse
Affiliation(s)
- Lakshi Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-39, Assam, India
| | - Sujisha S Nambiar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-39, Assam, India
| | - Arman Mohanty
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-39, Assam, India
| | - Arupam Patra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-39, Assam, India
| | - Shivani Kumari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-39, Assam, India
| | - Gurvinder Kaur Saini
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-39, Assam, India.
| |
Collapse
|
7
|
Wang X, Wang L, Yu J, Teng Y, Xiang X, Zhang D, Kang L, Niu Y, Feng X, Chen L. Effect of electron beam irradiation on the quality of chicken during post-mortem ageing. Food Chem 2025; 480:143869. [PMID: 40120307 DOI: 10.1016/j.foodchem.2025.143869] [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: 11/07/2024] [Revised: 02/15/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
The effects of electron beam irradiation (0, 4, and 8 kGy) on the quality of fresh chicken breast muscle during post-mortem ageing (0, 1, 3, and 5 days) were evaluated. The results suggested that the pH value and water-holding capacity of the chicken breast muscle were reduced, and the water was migrated. The color of the chicken breast muscle improved after irradiation, with decreased in L* and b* values and increased in a* values and oxymyoglobin content. Furthermore, irradiation significantly lowered shear force, raised the myofibrillar fragmentation index (MFI), and enhanced chicken breast tenderness. RT-qPCR and western blotting analyses showed that electron beam irradiation influenced the tenderness of chicken breast muscle by regulating apoptosis through mitochondrial, death receptor, and ERS pathways during post-mortem ageing. In conclusion, these results suggested that electron beam irradiation improved tenderness through apoptosis and changed chicken breast quality (such as color, pH, and moisture).
Collapse
Affiliation(s)
- Xin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Linya Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Jiangtao Yu
- Yangling Hesheng Irradiation Technologies Co., Ltd., Yangling 712100, China
| | - Yifeng Teng
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Xiaomei Xiang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Dan Zhang
- Yangling Hesheng Irradiation Technologies Co., Ltd., Yangling 712100, China
| | - Luyao Kang
- Yangling Hesheng Irradiation Technologies Co., Ltd., Yangling 712100, China
| | - Yabin Niu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Xianchao Feng
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Lin Chen
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
8
|
Palabiyik AA. The role of Bcl‑2 in controlling the transition between autophagy and apoptosis (Review). Mol Med Rep 2025; 32:172. [PMID: 40242969 PMCID: PMC12045647 DOI: 10.3892/mmr.2025.13537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
The Bcl‑2 protein family serves a key role in maintaining cellular homeostasis by regulating the balance between autophagy and apoptosis. The present review aimed to summarize interactions of Bcl‑2 with key proteins, including Beclin 1, Bax and Bcl‑2 homologous antagonist/killer, as well as its influence on cellular processes such as mitophagy, nutrient sensing and endoplasmic reticulum stress response. The impact of post‑translational modifications of Bcl‑2, including phosphorylation, ubiquitination and sumoylation, is discussed with respect to their regulatory roles under stress. In pathological states, Bcl‑2 upregulation in cancer suppresses apoptosis and autophagy, thereby facilitating tumor survival and resistance to chemotherapy. Conversely, in neurodegenerative diseases, impaired autophagy and increased apoptosis contribute to neuronal loss. Therapeutic strategies targeting Bcl‑2 (for example inhibitors such as venetoclax, navitoclax, obatoclax and combination therapies involving autophagy modulators) were evaluated for their potential efficacy. There is lack of understanding of tissue‑specific functions of Bcl‑2 and its interactions with non‑coding RNAs. Future research should prioritize these areas and leverage advanced single‑cell technologies to elucidate the real‑time dynamics of Bcl‑2 in cell processes. The present review highlights the key role of Bcl‑2 in cell fate determination and highlights its potential as a therapeutic target, offering insight for the development of innovative treatments for cancer, neurodegenerative disorder and age‑related diseases.
Collapse
Affiliation(s)
- Ahmet Alperen Palabiyik
- Department of Nursing, Faculty of Health Sciences, Ardahan University, Çamlıçatak, Ardahan 75002, Turkey
| |
Collapse
|
9
|
Pu P, Zheng F, Wang H, Wen H, Zhang L, Ma J. Integrating network pharmacology, experimental validation and molecular dynamics simulation to investigate the underlying mechanism of apigenin against diabetic wound. Biochem Biophys Res Commun 2025; 775:152113. [PMID: 40489928 DOI: 10.1016/j.bbrc.2025.152113] [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: 02/20/2025] [Revised: 05/20/2025] [Accepted: 05/28/2025] [Indexed: 06/11/2025]
Abstract
Diabetic wounds (DW) are one of the complications of diabetes. They are difficult to repair and current treatment options are limited. In this study, we employed a network pharmacology approach to investigate the therapeutic potential of Apigenin (API) in the treatment of DW. We used TCMSP, ETCM and SwissTargetPrediction to identify API's drug targets and DisGeNET, GeneCards, TTD, and OMIN for DW-related targets, yielding a set of intersecting genes. Then, we constructed the PPI network by STRING and visualized with Cytoscape software. Further refinement through relevant algorithms of Cytohubba identified nine core genes: HSP90AA1, ACTB, AKT1, BCL2, EGFR, SRC, CCND1, ESR1, and JUN. GO/KEGG analysis were conducted using the DAVID database. The results show that the targets are enriched in biological processes related to cell and smooth muscle movement and migration, as well as pathways associated with anti-inflammatory, diabetes, and inflammatory stimuli. Moreover, the binding affinity of API to identified core targets was confirmed through molecular docking using Autodock software. We further carried out Molecular docking simulations to verify that API exhibits significant binding affinities towards these core targets, suggesting a robust interaction profile. Finally, the experimental validation further confirmed the regulatory effect of API on PI3K-AKT signaling and cell apoptotic pathway. Our research elucidated that API holds significant promise as a therapeutic agent for the treatment of DWs.
Collapse
Affiliation(s)
- Pengpeng Pu
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai City, Hebei Province, 054001, China
| | - Fangjian Zheng
- Department of Orthopedic Surgery, Huang Jia Si Hospital, Jiangxi Province, 334700, China
| | - Huihui Wang
- Department of Anesthesiology, Hebei Eye Hospital, Hebei Treatment Center for Eye Disease, Xingtai City, Hebei Province, 050035, China
| | - Heli Wen
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province, 325035, China
| | - Liang Zhang
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, No.116 Zhuo Daoquan South Road, Wuhan, Hubei Province, 430079, China.
| | - Jie Ma
- Department of Cardiothoracic Surgery, Affiliated Hospital of Hubei University of Arts and Science, XiangYang, Hubei Province, 441000, China.
| |
Collapse
|
10
|
Imdad L, Xu S, Meng Y, Bao K, Dong W, Yin X, Tong Y, Zhang W, Ren X, Kong L. Txnip/Trx Is a Potential Element in Regulating O-GlcNAc Modification in Photoreceptors to Alleviate Diabetic Retinopathy. Int J Mol Sci 2025; 26:5369. [PMID: 40508179 PMCID: PMC12154564 DOI: 10.3390/ijms26115369] [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: 04/11/2025] [Revised: 05/14/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
Hyperglycemia is a key factor in diabetic retinopathy which leads to blindness. O-linked-N-acetylglucosamine (O-GlcNAc) modification changes are linked to various diseases, including diabetic retinopathy. This research aims to study the roles of Txnip and Trx in influencing O-GlcNAc in photoreceptor cells during diabetic retinopathy. A diabetic mouse model and 661w cells, after exposure to high glucose, were employed as models. H&E staining and ERG were used to evaluate the morphology and function of the retina, respectively. Western blotting was used to analyze protein expression, a TUNEL assay was used to measure apoptosis, and a co-immunoprecipitation (CO-IP) assay was used to detect the interactions of protein. In diabetic mice, electroretinogram (ERG) amplitude wave, retinal thickness, and body weight decreased. Glial fibrillary acidic protein (GFAP), Iba1 expression, and blood glucose level increased. In vitro, the percentage of apoptotic cell, Bax, and caspase3 levels increased, and Bcl2 decreased in 661w cells under high-glucose conditions. Moreover, Txnip expression was upregulated, while Trx was downregulated. Additionally, a Western blot analysis revealed that high-glucose exposure led to increased O-GlcNAc modification both in vivo and in vitro. The CO-IP results show that Txnip interacted with O-GlcNAc modifications. S-opsin expression was significantly downregulated in vitro under high-glucose conditions. Knockdown Txnip or upregulation Trx could reverse or delay apoptosis in 661w cells under hyperglycemic conditions. Txnip/Trx is a potential element in regulating photoreceptor apoptosis in diabetic retinopathy. The underlying mechanism is linked to regulation of O-GlcNAc modification in photoreceptor cells in DR.
Collapse
Affiliation(s)
- Laraib Imdad
- Department of Histology and Embryology, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Shengnan Xu
- Department of Histology and Embryology, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Yulang Meng
- Department of Histology and Embryology, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Kaimin Bao
- Department of Histology and Embryology, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Wenkang Dong
- Department of Histology and Embryology, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Xuanya Yin
- Department of Histology and Embryology, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Yujie Tong
- Department of Histology and Embryology, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Wei Zhang
- Department of Histology and Embryology, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Xiang Ren
- Department of Histology and Embryology, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
- Key Laboratory of Reproductive and Developmental Biology, Dalian Medical University, Dalian 116044, China
| | - Li Kong
- Department of Histology and Embryology, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
11
|
Gao F, Zhou X, Wei J, Sun Q, Wang J, Li Q. Expression characteristics and biological functions of CGB5 gene in gastric cancer. Acta Histochem 2025; 127:152254. [PMID: 40262449 DOI: 10.1016/j.acthis.2025.152254] [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: 10/17/2024] [Revised: 01/04/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVE The chorionic gonadotropin (CG) subunit beta 5 (CGB5) gene is a member of the glycoprotein hormone β chain family, encoding the β5 subunit of CG, which has been shown to promote tumorigenesis and induce proliferation in various types of cancer including gastric cancer (GC). However, the mechanistic role of CGB5 in GC has not been fully elucidated. Therefore, this study investigated relevant genes that regulate GC through bioinformatics analysis. METHODS Immunohistochemistry, immunofluorescence, and western blot (WB) detection methods were appropriately used to evaluate the expression pattern and clinical significance of CGB5 in 100 Chinese GC patients that were recruited from the Gaochun People's Hospital. The effect of small interfering ribonucleic acid (siRNA) on apoptosis, migration, and invasion of GC cells was investigated in vitro. Three-dimensional tumor spheres of these two types of GC cells (NCI-N87 cells and MKN45 cells) were constructed before investigation of the Calcein acetoxymethyl ester (AM)/ Propidium iodide (PI) staining, flow cytometric apoptosis, and apoptotic-related protein content of the tumor spheres after siRNA inhibition of CGB5 expression. RESULTS It was observed that compared with adjacent normal gastric tissue, expression of CGB5 was significantly upregulated in GC tissue. The siRNA inhibited CGB5 expression in two GC cell lines (NCI-N87 cells and MKN45 cells). Also, it was discovered that CGB5 highly correlated with microsatellite instability (MSI) and immune cell activity in GC, thus revealing the greater research value of CGB5 gene. More importantly, CGB5 siRNA could inhibit invasion and migration of tumor cells, induce apoptosis of GC cells and GC tumor spheres, as well as the mechanism relating to regulation of apoptosis associated gene expression. Overall, the findings suggest that CGB5 may play a crucial role in the development of GC carcinogenesis. Thus, this research may contribute to design of potential drug targets for treatment of GC.
Collapse
Affiliation(s)
- Fuping Gao
- Department of Pathology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaohua Zhou
- Department of Gastrointestinal Surgery, Gaochun People's Hospital, Nanjing, Jiangsu 211300, China
| | - Jin Wei
- Department of Pathology, Gaochun People's Hospital, Nanjing, Jiangsu 211300, China
| | - Qiong Sun
- Department of Pathology, Gaochun People's Hospital, Nanjing, Jiangsu 211300, China
| | - Jiapeng Wang
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Qing Li
- Department of Pathology, the Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
12
|
Kamala K, Ganapathy D, Sivaperumal P. Advancements in Cancer Therapy: Mycoviruses and Their Oncolytic Potential. Cell Biochem Biophys 2025; 83:1329-1332. [PMID: 39535660 DOI: 10.1007/s12013-024-01608-y] [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] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Recent advancements in cancer research focus on reducing treatment side effects while enhancing efficacy against medication resistance and tumor antigen detection. Genetic therapies utilizing microbes like bacteria, fungi, and viruses have garnered attention, with mycoviruses emerging as promising candidates. Particularly, the smallest fungal virus, Myco-phage, exhibits oncolytic properties by lysing cancer cells in the mouth, oral cavity, head, and neck without adverse effects. Genetically Modified Myco-phage (GmMP) adapts quickly to target cancer cells through cell membrane damage, inducing apoptosis and dendritic cell activation. Additionally, GmMP inhibits angiogenesis and modulates immune responses via CAR cells and immune checkpoints, potentially transforming cancer treatment paradigms with enhanced specificity and efficacy.
Collapse
Affiliation(s)
- Kannan Kamala
- Marine Microbial Research Lab, Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
- Centre for Marine and Aquatic Research (CMAR), Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 602105, India
| | - Dhanraj Ganapathy
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Pitchiah Sivaperumal
- Centre for Marine and Aquatic Research (CMAR), Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 602105, India.
- Marine Biomedical Research Lab & Environmental Toxicology Unit, Cellular and Molecular Research Centre, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India.
| |
Collapse
|
13
|
Chidambaram K, Rekha A, Goyal A, Rana M. Targeting KRAS-G12C in lung cancer: The emerging role of PROTACs in overcoming resistance. Pathol Res Pract 2025; 270:155954. [PMID: 40233529 DOI: 10.1016/j.prp.2025.155954] [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: 12/30/2024] [Revised: 03/30/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
In lung cancer, KRAS mutations, especially the G12C, favor aggressive tumor growth and resistance to standard therapies. Although first-generation inhibitors of KRAS G12C, such as sotorasib and adagrasib, are highly effective in early-phase studies, resistance invariably develops under selective inhibition pressure and rarely leads to sustained long-term treatment benefits. As a novel approach to targeting KRAS mutations in lung cancer, PROTAC (Proteolysis Targeting Chimera) technology is explored in this review. The PROTACs take advantage of the cell's ubiquitin-proteasome system to selectively degrade KRAS proteins, overcoming the dilemma of a lack of traditional binding sites and the means of resistance. We review recent progress with KRAS-specific PROTACs and their mechanisms, clinical application, and effectiveness at targeting primary KRAS oncogenes and secondary drivers and signaling pathways contributing to therapeutic resistance. Also, the synergies between PROTACs and immunotherapies or chemotherapies are further amplified. This review also underscores PROTAC technology's promise to advance precision medicine by providing durable treatment options for KRAS-driven lung cancers. It addresses future directions for optimizing PROTAC efficacy, bioavailability, and patient-specific applications.
Collapse
Affiliation(s)
- Kumarappan Chidambaram
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia.
| | - A Rekha
- Dr DY Patil Medical college , Hospital and Research Centre, Pimpri , Pune, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, UP 281406, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
14
|
Thakur B, Verma R, Bhatia A. Mutations in Necroptosis-Related Genes Reported in Breast Cancer: A Cosmic and Uniport Database-Based Study. Clin Breast Cancer 2025; 25:e341-e359. [PMID: 39794252 DOI: 10.1016/j.clbc.2024.12.008] [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: 05/18/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 01/13/2025]
Abstract
Breast cancer (BC) now holds the top position as the primary reason of cancer-related fatalities worldwide, overtaking lung cancer. BC is classified into diverse categories depending on histopathological type, hormone receptor status, and gene expression profile, with ongoing evolution in their classifications. Cancer initiates and advances when there is a disruption in cell death pathways. In BC, the primary cell death pathway, apoptosis, experiences dysregulation across multiple stages. Ongoing studies aim to discover therapeutic targets that enhance cancer cell susceptibility to apoptosis. However, resistance to this therapy remains a significant challenge in treating BC. If apoptosis is hindered, investigating alternative pathways for cell death that can effectively eradicate BC cells during treatment becomes a valuable endeavor. In this context, necroptosis is gaining considerable focus as an alternative cell death pathway. Necroptosis represents a programmed version of necrosis which shares its key regulators with apoptosis. When apoptosis is hampered, necroptosis serves as an alternative cell death pathway even in physiological conditions like formation of limbs during embryonic development. Additionally, it comes into play during bacterial and viral infections when the apoptosis machinery is hijacked and inhibited by proteins from these pathogens. Studies reveal that in BC, mutations significantly impact molecules in the apoptosis pathway, contributing to the onset, advancement, and multiplication of cancer cells. Although some studies do indicate that the functionality of necroptosis pathway may be compromised in malignancy the status of its key molecules remains largely unknown. In this article, we aim to gather the known mutations present in key molecules of necroptosis among various subtypes of BC, utilizing data from the Cosmic and UniProt databases. The same may help to enhance the development of therapeutic strategies to effectively induce necroptosis in apoptosis-resistant BCs.
Collapse
Affiliation(s)
- Banita Thakur
- Department of General Surgery, Stanford university, CA, USA
| | - Rohit Verma
- Department of Neurosurgery, Stanford University, CA, USA
| | - Alka Bhatia
- Department of Experimental Medicine & Biotechnology, PGIMER, Chandigarh, India.
| |
Collapse
|
15
|
Qin X, Wang X, Yang C, Wang F, Fang T, Gu D, Guo Q, Meng Q, Liu W, Yang L. A potent dual inhibitor targeting COX-2 and HDAC of acute myeloid leukemia cells. Mol Divers 2025; 29:2433-2444. [PMID: 39480610 DOI: 10.1007/s11030-024-11000-w] [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: 07/18/2024] [Accepted: 09/24/2024] [Indexed: 11/02/2024]
Abstract
Acute myeloid leukemia (AML) is an aggressive cancer with complex issues of drug resistance and a poor prognosis; thus, effective therapeutics is urgently needed for AML. In this study, we designed and synthesized dual cyclooxygenase-2 (COX-2) and histone deacetylase (HDAC) inhibitors, IMC-HA and IMC-OPD, and applied them for the treatment of AML. IMC-HA comprised a COX-2 inhibitor skeleton of indomethacin (IMC) and an HDAC inhibitor moiety of the hydroxamic group and was found to exhibit potent antiproliferative activity against AML cells (THP-1 and U937) and low cytotoxicity toward normal cells. Molecular docking simulations suggested that IMC-HA had a high binding affinity for HDAC and COX-2, with binding energies of -6.8 and -9.0 kcal/mol, respectively. Mechanistic studies revealed that IMC-HA induced apoptosis and G0/G1 phase arrest in AML cells, which were characterized by alterations in the expression of apoptotic and cell cycle-related proteins. Further study demonstrated that IMC-HA also inhibited the MEK/ERK signaling pathway in AML cells. Overall, we believe that IMC-HA could serve as a potent COX-2/HDAC dual inhibitor and improve the treatment of AML.
Collapse
Affiliation(s)
- Xiang Qin
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xueting Wang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Chunmei Yang
- Department of Radiology, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Fan Wang
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Tingting Fang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Didi Gu
- Department of Radiology, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qulian Guo
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qiuyu Meng
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Interdisciplinary Research Academy (IRA), Zhejiang Shuren University, Hangzhou, 310015, China.
| | - Wenjun Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Lu Yang
- Department of Radiology, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
16
|
He C, He J. Metabolic reprogramming and signaling adaptations in anoikis resistance: mechanisms and therapeutic targets. Mol Cell Biochem 2025; 480:3315-3342. [PMID: 39821582 DOI: 10.1007/s11010-024-05199-3] [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: 11/17/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025]
Abstract
Anoikis, a form of programmed cell death triggered by detachment from the extracellular matrix (ECM), maintains tissue homeostasis by removing mislocalized or detached cells. Cancer cells, however, have evolved multiple mechanisms to evade anoikis under conditions of ECM detachment, enabling survival and distant metastasis. Studies have identified differentially expressed proteins between suspended and adherent cancer cells, revealing that key metabolic and signaling pathways undergo significant alterations during the acquisition of anoikis resistance. This review explores the regulatory roles of epithelial-mesenchymal transition, cancer stem cell characteristics, metabolic reprogramming, and various signaling pathway alterations in promoting anoikis resistance. And the corresponding reagents and non-coding RNAs that target the aforementioned pathways are reviewed. By discussing the regulatory mechanisms that facilitate anoikis resistance in cancer cells, this review aims to shed light on potential strategies for inhibiting tumor progression and preventing metastasis.
Collapse
Affiliation(s)
- Chao He
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jie He
- Department of Nursing, Operating Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
17
|
Parveen S, Ahtsham A, Kumar S, Gupta P, Ghous F, Banerjee M. Piperlongumine mediated apoptosis in cervical cancer cells beyond docking predictions. Toxicol Rep 2025; 14:102031. [PMID: 40353245 PMCID: PMC12063131 DOI: 10.1016/j.toxrep.2025.102031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/01/2025] [Accepted: 04/16/2025] [Indexed: 05/14/2025] Open
Abstract
Cervical cancer is the 4th most prevalent cancer in women. Despite its global health impact, cervical cancer research has achieved limited success with various therapies (chemo, radio, chemo-radio, surgery, etc.). Recent decades have seen no improvement in survival rates due to cancer recurrence and long-term health concerns. These treatments show the need for a new herbal approach as alternative therapy for cancer. Piperlongumine is one of numerous perfected phytochemicals with anticancerous properties. In this study, in silico simulations demonstrate various novel properties of Piperlongumine. Molecular docking was performed between receptor and ligand by simulating molecular interactions in the pro and anti-apoptotic proteins B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax) and Bcl-2 antagonist/killer (Bak). The docking scores of Piperlongumine with Bcl-2, Bak, and Bax are -7.02, -6.78, and -7.54 kcal/mol, respectively. Piperlongumine was also bio-tested for anticancer activities against cervical cancer cells (SiHa). Piperlongumine showed compromised cell viability, promoted apoptosis, higher nuclear condensation, reduction in mitochondrial membrane potential, increase in reactive oxygen species level, promoted cell cycle arrest, upregulation of pro-apoptotic gene whereas downregulation of anti-apoptotic genes. In vitro studies revealed that Piperlongumine targets apoptotic pathway in cervical cancer and could be a possible effective treatment for cervical cancer. Additional in vivo research is needed to explore the potential of Piperlongumine to improve treatment outcomes.
Collapse
Affiliation(s)
- Shama Parveen
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Ana Ahtsham
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Saurabh Kumar
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Pratishtha Gupta
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Faraz Ghous
- Department of Chemistry, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Monisha Banerjee
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| |
Collapse
|
18
|
Wu X, Ali SSF, Jiang S, Zhong Y, Xu Y, Wei L, Zhang S, Feng Z, Huang X, Shi X, Mu Y, Wang X, Gan C, Yang C. Ultrasound-assisted combined with natural deep eutectic solvents for Platycodon grandiflorum polysaccharides extraction: Process optimization and evaluation of anti-lung cancer activity. Int J Biol Macromol 2025; 313:144190. [PMID: 40373906 DOI: 10.1016/j.ijbiomac.2025.144190] [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: 02/28/2025] [Revised: 04/21/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Platycodon grandiflorum (PG) is a traditional medicinal herb with a history of >2000 years, exhibiting various biological activities. However, its anti-lung cancer potential remains largely unexplored. This study aimed to extract Platycodon grandiflorum polysaccharides (PGP) using an environmentally friendly natural deep eutectic solvent (NADES) system and evaluate their anti-lung cancer activity. An NADES system composed of choline chloride and oxalic acid (1:3 M ratio) was selected based on carbohydrate yield. The extraction conditions were optimized using Box-Behnken design (BBD). The structural characterization of PGP was performed using ultraviolet spectroscopy, fourier transform infrared (FT-IR) spectroscopy, and high-performance liquid chromatography (HPLC), revealing that PGP are glucans with a molecular weight of 7.036 kDa and a glucose content of 92.8 %. Biological assays demonstrated that PGP promoted ferroptosis in H1299 cells through mitochondrial dysfunction, lipid peroxidation (LPO), and overproduction of reactive oxygen species (ROS). Also, PGP inhibited cell migration by modulating epithelial-mesenchymal transition(EMT)-related protein expression. These findings provide novel insights into the pharmacological basis of PG and support its potential application in lung cancer therapy.
Collapse
Affiliation(s)
- Xiaotian Wu
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Shah Syed Faizan Ali
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Shuang Jiang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Yinze Zhong
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Yeqing Xu
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Lin Wei
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Shulu Zhang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Zheng Feng
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Xinhui Huang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Xuepeng Shi
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Yuanqiu Mu
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Xiaotong Wang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Chunli Gan
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Chunjuan Yang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China.
| |
Collapse
|
19
|
Yadav N, Xiao A, Zhong Q, Kumar P, Konduru G, Hart W, Lazzara M, Purow B. Synergistic activity of simvastatin and irinotecan chemotherapy against glioblastoma converges on TGF-β signaling. J Neurooncol 2025:10.1007/s11060-025-05089-8. [PMID: 40434539 DOI: 10.1007/s11060-025-05089-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025]
Abstract
PURPOSE This study investigates the synergistic therapeutic potential of a novel combination of the repurposed drug simvastatin with irinotecan chemotherapy towards glioblastoma (GBM) and the underlying molecular mechanisms. METHODS In vitro efficacy of simvastatin and irinotecan alone and in combination against diverse GBM lines (U251MG, G34, SB28) was assessed using mechanistically distinct cell viability assays. RNA-Sequencing was performed to uncover the top pathways and genes affected by these drugs, followed by validation of promising pathways (TGF-β signaling and cell death) using targeted phosphoproteomics and in vitro genetic manipulation and functional assays. RESULTS We observed robust in vitro synergy at nanomolar concentrations between simvastatin and irinotecan across diverse GBM lines. Notably, irinotecan alone and in combination with simvastatin reduced mRNA expression of TGF-β family members. Targeted phosphoproteomics and functional experiments further showed significant inhibition of TGF-β signaling with both treatment types. Additionally, a role for apoptosis and enrichment of caspase-independent cell death pathways (autophagy, ferroptosis) as well as immunological (interferons, complement, inflammatory responses, TNF-α) and oncogenic (K-RAS/ERK) signaling pathways were observed with the combination treatment. CONCLUSIONS Besides the first detailed demonstration of a robust synergy between simvastatin and irinotecan against GBM lines, this study shows for the first time that both irinotecan and the combination treatment converge on inhibition of TGF-β signaling. This is notable given the lack of TGF-β inhibitors in the clinic. Collectively, this study provides preclinical data suggesting this novel drug combination be tested in patients with GBM and TGF-β driven cancers.
Collapse
Affiliation(s)
- Niket Yadav
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
- Medical Scientist Training Program (MSTP), University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
- Molecular and Cellular Basis of Disease (MCBD) Graduate Program, Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Aizhen Xiao
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Qing Zhong
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Pankaj Kumar
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA
- Bioinformatics Core, University of Virginia, Charlottesville, VA, 22903, USA
| | - Guruprasad Konduru
- Bioinformatics Core, University of Virginia, Charlottesville, VA, 22903, USA
| | - William Hart
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
| | - Matthew Lazzara
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
| | - Benjamin Purow
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA.
| |
Collapse
|
20
|
Sarı ZB, Sarı ME, Aytar EC, Gümrükçüoğlu A, Torunoğlu EI, Ozdemir-Sanci T, Demirel G, Durmaz A. Flow Cytometry and Gene Expression Modulation by Euphorbia rigida Methanol Extract in A549 Lung Cancer Cells: Induction of Apoptosis Through Bax, Caspase-9, and Bcl-2 Pathways. Adv Biol (Weinh) 2025:e00136. [PMID: 40432491 DOI: 10.1002/adbi.202500136] [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: 02/13/2025] [Revised: 05/06/2025] [Indexed: 05/29/2025]
Abstract
Non-small cell lung cancer (NSCLC) remains a major cause of cancer-related mortality. This study investigates the cytotoxic effects of Euphorbia rigida extract on A549 NSCLC cells and its potential as a therapeutic agent. Cellular morphology was observed microscopically, and cell viability was evaluated using dose-dependent proliferation assays. Apoptosis-related gene expression-including Bax, Bcl-2, and Caspase-9-was analyzed via quantitative PCR (qPCR). Chromatographic methods identified bioactive flavonoids, and molecular docking assessed their binding to cancer-related proteins. Additionally, absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiles were evaluated. The extract induced apoptotic morphological changes such as cell shrinkage and loss of intercellular contact. A dose-dependent reduction in A549 viability was observed, with an IC50 of 0.5 mg mL-1. Gene expression indicated activation of the intrinsic mitochondrial apoptotic pathway, with increased Bax and Caspase-9 and decreased Bcl-2 expression. Flow cytometry using Annexin V-allophycocyanin (V-APC) staining revealed selective cytotoxicity: significant apoptosis in A549 cells while preserving viability in BEAS-2B normal lung epithelial cells. Identified flavonoids included quercetin, apigenin, and myricetin, which showed strong binding affinities in docking studies. ADMET profiling supported their drug-likeness. These findings highlight E. rigida potential in NSCLC treatment via apoptosis induction and selective cytotoxicity.
Collapse
Affiliation(s)
- Zeynep Betül Sarı
- Faculty of Medicine, Department of Basic Medical Sciences, Medical Biology, Ankara Yıldırım Beyazıt University, Ankara, 06010, Türkiye
| | - Muhammed Emin Sarı
- Faculty of Medicine, Department of Medical Biology, Necmettin Erbakan University, Konya, 42090, Türkiye
| | - Erdi Can Aytar
- Faculty of Agriculture, Department of Horticulture, Usak University, Uşak, 64200, Türkiye
| | - Abidin Gümrükçüoğlu
- Medicinal-Aromatic Plants Application and Research Center, Artvin Çoruh University, Artvin, 08000, Türkiye
| | - Emine Incilay Torunoğlu
- Faculty of Medicine, Department of Medical Biochemistry, Necmettin Erbakan University, Konya, 42090, Türkiye
| | - Tuba Ozdemir-Sanci
- Faculty of Medicine, Department of Histology and Embriyology, Ankara Yildirim Beyazit University, Ankara, 06010, Türkiye
| | - Gamze Demirel
- Selçuk University - Akşehir Kadir Yallagöz School of Health - Department of Nutrition and Dietetics, Konya, 42560, Türkiye
| | - Alper Durmaz
- Ali Nihat Gökyiğit Botanical Garden Application and Research Center, Artvin Çoruh University, Artvin, 08000, Türkiye
| |
Collapse
|
21
|
Velmurugan BK, Lin CC, Kao MY, Ho HY, Lo YS, Chuang YC, Hsieh MJ. Limocitrin induced cellular death through ERK pathways in human oral squamous cell cancer. Sci Rep 2025; 15:17788. [PMID: 40404781 PMCID: PMC12098972 DOI: 10.1038/s41598-025-02178-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 05/12/2025] [Indexed: 05/24/2025] Open
Abstract
This study sought to investigate the anticancer efficacy of limocitrin on two distinct human oral cancer cell lines. At first, we evaluated the effect of limocitrin on the proliferation of OSCC cells (SCC-9 and SCC-47) using MTT and colony formation assays. Limocitrin treatment increased cell cycle arrest at G2/M phase, induced caspase-related apoptosis (cleaved caspase-3, caspase-8, caspase-9 and PARP expression) in OSCC cells. Limocitrin treatment inhibited Bcl-2 and Bcl-XL and induced Bax, Bak expression in both SCC-9 and SCC-47 cell lines. Limocitrin treatment inhibited cyclin E1, E2, CDK2, CDK4, and CDK6 and increased p21 expression. Limocitrin also exhibited an inhibitory effect on the phosphorylation of AKT, ERK1/2 and JNK in a dose-dependent manner. Additionally, pretreatment of oral cancer cells with U0126 resulted in increased cleaved caspase-3 and caspase-8 and PARP expression. Furthermore, limocitrin treatment decreased XIAP, cIAP1, HSP27 protein expression than control group. Combined treatment with limocitrin and si-XIAP significantly increased cleaved PARP, caspase-3 and -8 expressions in SCC-9 cells. Overall, this evidence indicates that limocitrin may serve as an effective anticancer agent for the treatment of oral cancer.
Collapse
Affiliation(s)
| | - Chia-Chieh Lin
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 50006, Taiwan
| | - Min-Yun Kao
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 50006, Taiwan
| | - Hsin-Yu Ho
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 50006, Taiwan
| | - Yu-Sheng Lo
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 50006, Taiwan
| | - Yi-Ching Chuang
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 50006, Taiwan
| | - Ming-Ju Hsieh
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 50006, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, National Chung Hsing University, Taichung, 402202, Taiwan.
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, 402, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan.
| |
Collapse
|
22
|
Hu D, Chen K. Construction of a circRNA-miRNA-mRNA regulatory network in glioblastoma multiforme based on bioinformatics analysis. Medicine (Baltimore) 2025; 104:e42392. [PMID: 40355207 PMCID: PMC12074040 DOI: 10.1097/md.0000000000042392] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 04/18/2025] [Indexed: 05/14/2025] Open
Abstract
This study aimed to investigate the functional roles and molecular regulatory mechanisms of circular RNAs in the development of glioblastoma multiforme. Differentially expressed circular RNAs were identified by integrating RNA sequencing data and circular RNA microarray data from the Gene Expression Omnibus database. CircAtlas and CircInteractome databases were used to predict microRNAs (miRNAs) interacting with these circular RNAs. Survival analysis of the miRNAs was performed using data from the Chinese Glioma Genome Atlas. The miRTarBase database was used to predict miRNA target genes, followed by the construction of a circular RNA-miRNA-messenger RNA regulatory network specific to glioblastoma multiforme. Functional enrichment analysis was carried out using the DAVID website, and protein-protein interaction networks were created using the Search Tool for the Retrieval of Interacting Genes/Proteins website and Cytoscape. Hub genes were identified, and their expression and prognostic relevance in glioblastoma multiforme were further examined. Four differentially expressed circRNAs and 10 associated miRNAs related to glioblastoma multiforme prognosis were identified. Functional enrichment showed the miRNAs target genes were mainly involved in apoptosis, cell cycle regulation and enriched in cancer-related pathways like mitogen-activated protein kinase and PI3K-Akt. Through the circRNA-miRNA-messenger RNA regulatory network and survival analysis, 3 core genes (core hub genes: catenin beta 1, BCL2, nuclear factor kappa B subunit 1) were identified as significantly downregulated in glioblastoma multiforme and associated with patient survival. This study highlights the potential regulatory roles of circular RNAs in glioblastoma multiforme pathogenesis and their involvement in key molecular pathways. The findings offer a theoretical foundation for understanding glioblastoma multiforme development and may facilitate the identification of novel biomarkers for this aggressive cancer.
Collapse
Affiliation(s)
- Dongpo Hu
- School of Medical Technology, Shangqiu Medical College, Shangqiu, Henan, China
| | - Kangjing Chen
- School of Medical Technology, Shangqiu Medical College, Shangqiu, Henan, China
| |
Collapse
|
23
|
Hasan R, Hasan MM, Shathi JH, Tamam E, Ahmed AE, Haque A, Rahmann Z, Islam MT, Reza MA, Biswas MS, Hoque KMF. Toxic effects of malachite green on plant and animal models: A study on root growth inhibition, hematological changes, histopathology, and molecular analysis. Toxicol Lett 2025; 409:61-73. [PMID: 40348348 DOI: 10.1016/j.toxlet.2025.05.003] [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: 10/22/2024] [Revised: 04/03/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Malachite green (MG), a suggestive chemical for tumor development and carcinogenicity, is widely used as an illicit food coloring agent, posing risks to consumers and handlers. This study aimed to assess the toxic effects of MG in both plant and animal models. Different doses of MG (375, 750, and 1500 mg/L) were applied for 24 h to evaluate root growth inhibition, mitotic index (MI), and chromosomal aberrations in Allium cepa L. roots for genotoxicity analysis. In animal studies, forty Swiss albino mice were divided into four groups: control and three treatment groups, which were orally administered MG at 375 (low), 750 (medium), and 1500 (high) mg/kg body weight for 13 weeks. Hematological, biochemical, histopathological, and molecular analyses were performed on liver, kidney, and intestinal tissues post-treatment. MG significantly reduced root length and MI in A. cepa roots dose-dependently causing chromosomal abnormalities. MG treatment significantly lowered the body weights of mice and increased platelet, monocyte, and white blood cell counts, while reducing hemoglobin, hematocrit, and red blood cell counts. Serum analysis showed elevated ALT, ALP, AST, bilirubin, creatinine, and urea, indicating hepatotoxicity and nephrotoxicity. Histopathological examination revealed vacuolation, congestion, and inflammatory infiltration in the liver, glomerular shrinkage, tubular degeneration, and interstitial edema in the kidney, and epithelial sloughing, submucosal necrosis, and inflammatory infiltration in the colon. RT-qPCR analysis demonstrated increased Bcl-2, Beclin-1, and NF-κB mRNA expression with decreased Bax mRNA. These findings suggest MG is a potent genotoxic and carcinogenic agent even at lower doses threatening human health.
Collapse
Affiliation(s)
- Rubait Hasan
- Department of Biochemistry & Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh; Molecular Biology and Protein Science Laboratory, Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Mahmudul Hasan
- Molecular Biology and Protein Science Laboratory, Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Jamiatul Husna Shathi
- Department of Biochemistry & Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh; Molecular Biology and Protein Science Laboratory, Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Esraa Tamam
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Egypt
| | - Amaal E Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Aswan University, Egypt
| | - Ariful Haque
- Molecular Pathology Laboratory, Institute of Biological Sciences, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Zahidur Rahmann
- Department of Biochemistry & Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh; Riceland Healthcare, 538 Broadway Ave, Winnie, TX 77665, USA
| | - Md Tariqul Islam
- Department of Biochemistry & Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
| | - Md Abu Reza
- Molecular Biology and Protein Science Laboratory, Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Mohammad Shahangir Biswas
- Department of Biochemistry and Biotechnology, University of Science & Technology Chittagong (USTC), Foy's Lake, Zakir Hossain Road, Chittagong 4202, Bangladesh; Department of Public Health, Daffodil International University, Dhaka 1216, Bangladesh.
| | - Kazi Md Faisal Hoque
- Molecular Biology and Protein Science Laboratory, Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh.
| |
Collapse
|
24
|
Potdar MB, Bhamare RP, Agrawal YO, Belgamwar AV. Crosstalk Between Signaling Stroke Cascade and Therapeutic Receptors PPAR-γ, ROCK, CB1R, and CB2R: From Mechanism to Therapies. Transl Stroke Res 2025:10.1007/s12975-025-01352-2. [PMID: 40338418 DOI: 10.1007/s12975-025-01352-2] [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: 12/14/2024] [Revised: 02/07/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025]
Abstract
Stroke remains a leading cause of disability and mortality worldwide, primarily due to the complex and multifaceted nature of its pathophysiology. This review aims to provide a comprehensive and mechanistic understanding of the crosstalk between key signaling pathways activated during stroke and the therapeutic potential of specific receptors: PPAR-γ, ROCK, CB1R, and CB2R. We delve into the intricate signaling cascades that occur post-stroke, including excitotoxicity, oxidative stress, and inflammation, highlighting the pivotal molecular players involved. PPAR-γ, known for its neuroprotective and anti-inflammatory properties, emerges as a critical modulator in stroke therapy. ROCK, a central component in the Rho/ROCK pathway, is implicated in vascular and neuronal damage, making its inhibition a promising therapeutic strategy. The roles of CB1R and CB2R within the endocannabinoid system are explored, with a focus on their dualistic nature in neuroprotection and neurotoxicity. The review further examines the interconnectivity of these receptors within the stroke signaling network, proposing that their synergistic modulation could enhance therapeutic outcomes. Current therapeutic approaches, including pharmacological and multi-target strategies, are critically evaluated, addressing the challenges in translating mechanistic insights into clinical practice. Additionally, the identification and utilization of biomarkers for stroke diagnosis and therapy monitoring are discussed, offering a glimpse into future prospects. Emerging therapies, novel drug developments, and personalized medicine approaches are presented as potential game-changers in stroke treatment.
Collapse
Affiliation(s)
- Mrugendra B Potdar
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Rohit P Bhamare
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Yogeeta O Agrawal
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Aarti V Belgamwar
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India.
| |
Collapse
|
25
|
Lemos IL, Macedo MJ, Machado APDF, do Nascimento RDP, Reguengo LM, Cagnon VHA, Junior MRM. The chemopreventive effects of native Brazilian plants on stomach cancer: A review of the last 25 years. Oncoscience 2025; 12:36-51. [PMID: 40343252 PMCID: PMC12060929 DOI: 10.18632/oncoscience.618] [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: 03/11/2025] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
Stomach cancer (SC) is the fifth most prevalent and deathly type of cancer worldwide. This is a multifactorial disease, and its development can be influenced by both genetic factors and dietary habits. On the other hand, a regular consumption of fruit and vegetables rich in bioactive compounds, such as polyphenols and flavonoids, has demonstrated anti-inflammatory, antioxidant, and chemopreventive effects on SC. Brazil, which has a vast plant diversity, appears to be a promising scenario for investigating species with potential anti-tumor action. Thus, the objective of this review is to present and discuss the chemopreventive aspects of native Brazilian species in SC. Less-explored fractions of native plants, such as açaí (Euterpe oleracea), araçá-do-campo (Psidium guineense), yellow araçá (Psidium cattleianum Sabine), cacao (Theobroma cacao), coriander (Eryngium foetidum), physalis (Physalis angulata), guava (Psidium guajava), jambu (Acmella oleracea), pitanga (Eugenia uniflora), and ubaia (Eugenia patrisii), have demonstrated the ability to slow down the progression of the disease, indicating suppression of cell proliferation and survival, induction of apoptosis, and regulation of the cell cycle, despite showing not mechanism of action in the great majority of these studies. Although, still little studied, Brazilian plant matrices could show a promising impact against SC.
Collapse
Affiliation(s)
- Iara Lopes Lemos
- Laboratory of Nutrition and Metabolism, School of Food Engineering, University of Campinas, Campinas 13083-862, São Paulo, Brazil
| | - Maria Josiane Macedo
- Laboratory of Nutrition and Metabolism, School of Food Engineering, University of Campinas, Campinas 13083-862, São Paulo, Brazil
| | - Ana Paula da Fonseca Machado
- Laboratory of Nutrition and Metabolism, School of Food Engineering, University of Campinas, Campinas 13083-862, São Paulo, Brazil
| | - Roberto de Paula do Nascimento
- Laboratory of Nutrition and Metabolism, School of Food Engineering, University of Campinas, Campinas 13083-862, São Paulo, Brazil
| | - Lívia Mateus Reguengo
- Laboratory of Nutrition and Metabolism, School of Food Engineering, University of Campinas, Campinas 13083-862, São Paulo, Brazil
| | - Valeria Helena Alves Cagnon
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP) Campinas 13083-862, São Paulo, Brazil
| | - Mario Roberto Marostica Junior
- Laboratory of Nutrition and Metabolism, School of Food Engineering, University of Campinas, Campinas 13083-862, São Paulo, Brazil
| |
Collapse
|
26
|
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
|
27
|
Abdelhamid SA, Mohamed SS, Abo Elsoud MM, Selim MS, Mounier MM, Eltaher A, Magdeldin S, Ali M, Awady MEE. Characterization and Modeling of Marine Bacillus cereus Strain MSS1 Exopolysaccharide and Its Antagonistic Effect on Colon Cancer. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10539-w. [PMID: 40320507 DOI: 10.1007/s12602-025-10539-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2025] [Indexed: 05/17/2025]
Abstract
Microbial polysaccharides are a significant group of functional phytochemicals. Numerous studies have shown the advantageous pharmacological impacts of polysaccharides, including their effectiveness against cancer. A halophilic bacterial strain obtained from coastal sediments produced exopolysaccharides (EPS). The strain was morphologically recognized and further confirmed as Bacillus cereus strain MSS1 using 16S rDNA analysis, with accession number OR133726. The heteropolysaccharides were purified and fractionated with a DEAE-cellulose column, and the preliminary chemical analysis of the most potent fraction (EPSMSS1) indicated that the four different monosaccharides were mannuronic acid, xylose, fructose, and glucuronic acid, with a molar ratio of 1:1:2:0.5, respectively. The highest production was 12.76 g/l using a Box-Behnken design. It showed antibacterial activity, antioxidant activity, and antibiofilm activity. The 3D architecture of the EPSMSS1 of Bacillus cereus strain MSS1 is being described, predicted, and aligned against other bacterial species. These studies offer valuable insights into optimizing efficiency. Therefore, the EPSMSS1 fraction was shown to have anticancer activity and significant anticancer activities in a dose-dependent manner, with an IC50 value of 20.1 µg/ml. Subsequently, various apoptotic markers, such as cytochrome c, BAX, BCl2, and the BAX/BCL2 ratio, were assessed. Our findings demonstrate that EPSMSS1 triggers the activation of apoptotic protein BAX, enhances the generation of cytochrome c, reduces the expression of antiapoptotic protein BCl2, and distorts the BAX/BCL2 ratio in EPSMSS1-treated HCT-116 cells relative to untreated cells. The anticancer efficacy of EPSMSS1 was verified through the assessment of cell cycle progression using flow cytometry. It has been found that EPSMSS1 pauses the cell cycle in the G1/S phase, causing apoptosis. The main motivation behind this study was EPSMSS1, an innovative marine polysaccharide with remarkable biological activity, especially anti-cancer properties.
Collapse
Affiliation(s)
| | - Sahar S Mohamed
- Microbial Biotechnology Department, National Research Centre, Cairo, Egypt
| | | | - Manal S Selim
- Microbial Biotechnology Department, National Research Centre, Cairo, Egypt
| | - Marwa M Mounier
- Pharmacognosy Department, National Research Centre, Cairo, Egypt
| | - Ashraf Eltaher
- Proteomics and Metabolomics Research Program, Research Department, Basic Research Unit, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
| | - Sameh Magdeldin
- Proteomics and Metabolomics Research Program, Research Department, Basic Research Unit, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Mohamed Ali
- Biochemistry Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed E El Awady
- Microbial Biotechnology Department, National Research Centre, Cairo, Egypt
| |
Collapse
|
28
|
Khan SK, Dutta J, Rather MA, Ahmad I, Nazir J, Karnwal A. Toxicological impact of copper nanoparticles on rainbow trout: hematological, biochemical, antioxidant, and histopathological responses with oxidative gene expression. Toxicol Mech Methods 2025; 35:356-371. [PMID: 39654112 DOI: 10.1080/15376516.2024.2438120] [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: 10/06/2024] [Revised: 11/23/2024] [Accepted: 11/29/2024] [Indexed: 05/04/2025]
Abstract
Copper nanoparticles (CuNPs) are increasingly used across various industries due to their catalytic, antimicrobial, and electrical properties. However, their potential toxicity in aquatic environments, particularly to non-target organisms like fish, remains poorly understood. This study investigated the effects of CuNP exposure on rainbow trout (Oncorhynchus mykiss) by comprehensively evaluating hematological, biochemical, antioxidant, molecular, and histopathological parameters. Rainbow trout fingerlings were exposed to varying concentrations of CuNPs (0.2 mg/L, 0.6 mg/L, and 1.0 mg/L) for 7, 14, and 21 days. The results revealed significant dose-dependent declines in hemoglobin (Hb) and red blood cell (RBC) counts, alongside increases in white blood cell (WBC) counts, indicating an immune response to CuNPs-induced stress. Serum biochemistry showed disruptions in albumin, globulin, cholesterol, and triglycerides, suggesting impaired liver function and altered lipid metabolism. Antioxidant enzyme activity, including catalase (CAT), increased significantly, reflecting oxidative stress, while lipid peroxidation (LPO) levels unexpectedly decreased, suggesting possible activation of compensatory mechanisms. Histopathological analysis confirmed severe gill and liver damage, including hypertrophy, hyperplasia, lamellar fusion, necrosis, and cellular degeneration. Molecular analysis showed upregulation of oxidative and, inflammatory genes, and signs of apoptosis. These findings underscore the toxic potential of CuNPs in aquatic environments and highlight the need for careful regulation and environmental monitoring to mitigate their impact.
Collapse
Affiliation(s)
- Saba Khursheed Khan
- Department of Zoology, School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Joydeep Dutta
- Department of Zoology, School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Mohd Ashraf Rather
- Division of Fish Genetics and Biotechnology, Faculty of Fisheries Ganderbal, Sher-e- Kashmir University of Agricultural Science and Technology, Kashmir, India
| | - Ishtiyaq Ahmad
- Division of Fish Genetics and Biotechnology, Faculty of Fisheries Ganderbal, Sher-e- Kashmir University of Agricultural Science and Technology, Kashmir, India
| | - Junaid Nazir
- Department of Clinical biochemistry, Lovely Professional University, Phagwara, Punjab, India
| | - Arun Karnwal
- Department of Microbiology, Graphic Era (Deemed to be University), Dehradun, Uttarrakhand, India
| |
Collapse
|
29
|
Ren G, Lin Y, Fu Y, Liu F, Wang R, Zhang C, Qiu J, Chen L, Dou X. Multi-omics joint analysis: Pachymic acid ameliorated non-alcoholic fatty liver disease by regulating gut microbiota. Food Res Int 2025; 209:116178. [PMID: 40253122 DOI: 10.1016/j.foodres.2025.116178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/12/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Poria cocos a traditional Chinese medicinal material with both culinary and therapeutic applications, contains pachymic acid (Pac) as one of its main active compounds, which has demonstrated anti-lipid accumulation and hypoglycemic effects. However, its impact on the biochemical changes in the enterohepatic axis induced by a high-fat diet remains poorly understood. This study investigated the protective mechanism of Pac using a high-fat diet-induced non-alcoholic fatty liver disease (NAFLD) mouse model. 16S rRNA sequencing of gut microbiota revealed that Pac administration reduced the Firmicutes to Bacteroidetes ratio, restored Akkermansia abundance, decreased Desulfovibrio and Streptococcus population, and ameliorated gut dysbiosis. Concurrently, Pac treatment reduced the expression of hepatic inflammatory factors by mainly adjusted LPS/TLR4/MYD88/NFκB pathway. Liver transcriptome analysis indicated that Pac primarily affects genes involved in lipid metabolism, apoptosis, and inflammatory responses. Specifically, Pac inhibited FASN, SREBP1c, and SCD1 expression while upregulating PPARα and CPT1α, thereby improving high-fat diet-induced hepatic steatosis in mice. Additionally, Pac treatment reduced hepatocellular apoptosis. Non-targeted liver metabolomics analysis following Pac intervention revealed increased levels of acylcarnitine and oleic acid. Collectively, these findings suggest that Pac alleviates high-fat diet-induced hepatic lipid accumulation and damage by modulating gut microbiota, lipid metabolism, inflammation, and apoptosis. This comprehensive study provides valuable insights into the therapeutic potential of Pac and offers a reference for the development and utilization of Poria cocos resources in addressing NAFLD.
Collapse
Affiliation(s)
- Guilin Ren
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yiyou Lin
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yilong Fu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Fucai Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Ruiqi Wang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Congcong Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Jiannan Qiu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Lin Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
30
|
Saleh DO, Abo El Nasr NME, Hussien YA, El-Baset MA, Ahmed KA. Cyclophosphamide-induced testicular injury: the role of chrysin in mitigating iron overload and ferroptosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5475-5489. [PMID: 39565397 PMCID: PMC11985597 DOI: 10.1007/s00210-024-03519-4] [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: 07/28/2024] [Accepted: 10/06/2024] [Indexed: 11/21/2024]
Abstract
This study evaluated the beneficial effects of chrysin against cyclophosphamide (CP)-induced testicular toxicity in rats across several parameters, including hormones, oxidative stress, inflammation, apoptosis, and protein expression. Rats were pretreated with oral doses of chrysin at 25, 50, or 100 mg/kg daily for 7 days. On the 8th day, all groups except controls received CP (200 mg/kg) injection. Chrysin doses continued for 7 more days. Hormones, oxidative stress markers, inflammatory cytokines, apoptosis regulators, and iron regulatory proteins were assessed. CP decreased testosterone, inhibin B, GSH, and GPx4 and increased FSH, cholesterol, MDA, IL-6, and BAX. It also drastically reduced TfR1, liprin, and IREB2. Chrysin dose-dependently counteracted these effects. The highest 100 mg/kg chrysin dose increased testosterone, inhibin B, GSH, GPx4, BCL2, TfR1, liprin, and IREB2 while decreasing FSH, cholesterol, MDA, IL-6, and BAX close to control levels. There were also significant incremental benefits for testosterone, inhibin B, and other parameters with higher chrysin doses. Chrysin dose-dependently attenuated CP-induced hormonal dysfunction, oxidative stress, inflammation, apoptosis, and iron-regulatory protein suppression. The maximum dose showed the most optimal protective effects in restoring the testicular toxicity markers. These results validate the promising spermatoprotective properties of chrysin against chemotherapeutic germ cell damage.
Collapse
Affiliation(s)
- Dalia O Saleh
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Nesma M E Abo El Nasr
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt.
| | - Yosra A Hussien
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Marawan Abd El-Baset
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Kawkab A Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
31
|
Ding W, Wang L, Wu J, Li H, Li B, Wang Y, Zhang J, Li W. Neuroprotective effects of macrostemonoside T on glutamate-induced injury in HT22 cells. Biochem Pharmacol 2025; 235:116827. [PMID: 39993610 DOI: 10.1016/j.bcp.2025.116827] [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: 10/02/2024] [Revised: 02/12/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
Glutamate is a critical excitatory neurotransmitter involved in numerous cellular functions. However, excessive glutamate release can lead to neuronal cell death through oxidative stress, which is implicated in the pathogenesis of various neurological disorders. Therefore, strategies aimed at preventing oxidative stress have emerged as promising therapeutic approaches. Macrostemonoside T (MST), a novel steroidal saponin isolated from the traditional Chinese medicine Allii Macrostemon Bulbus, has demonstrated significant antioxidant activity in previous studies. Nevertheless, its neuroprotective effects against oxidative damage and the underlying molecular mechanisms have not yet been fully elucidated. In this study, we established a glutamate-induced cell injury model using mouse hippocampal neurons (HT22) to investigate the neuroprotective effects of MST and explore its potential mechanisms. A variety of techniques, including DCFH-DA staining, JC-1 staining, Hoechst 33,258 staining, flow cytometry, immunofluorescence staining, ELISA, Western blot analysis, and molecular docking, were employed. The results demonstrated that MST treatment significantly improved the survival of HT22 cells exposed to glutamate. Moreover, MST treatment markedly reduced intracellular levels of reactive oxygen species (ROS) and malondialdehyde while enhancing the activity of antioxidant enzymes such as superoxide dismutase, catalase, and glutathione peroxidase. MST also mitigated mitochondrial dysfunction by inhibiting mitochondrial fission and preserving mitochondrial membrane potential. Additionally, MST reduced excessive autophagy by decreasing autophagy markers and inhibiting the transition from LC3I to LC3II. Furthermore, MST decreased apoptosis rates, lowered pro-apoptotic protein BAX levels, increased the expression of the anti-apoptotic protein Bcl-2, and inhibited the release of apoptosis-inducing factors from mitochondria. Molecular docking analysis showed that MST enhanced PKA activity by blocking endogenous inhibition of PKA, which in turn activated the PKA/CREB/BDNF signalling pathway. Subsequent validation using immunofluorescence and Western blotting further confirmed that MST treatment significantly reversed the glutamate-induced reduction of PRKACA, CREB, p-CREB, and BDNF protein levels. In conclusion, MST is a potent neuroprotective agent that ameliorates glutamate-induced neuronal damage by inhibiting oxidative stress, alleviating mitochondrial dysfunction, reducing autophagy and apoptosis, and activating the PKA/CREB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Weixing Ding
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Lulu Wang
- College of Medicine, Changchun Sci-Tech University, Changchun 130600, China
| | - Jianfa Wu
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Hongyan Li
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Bing Li
- College of Medicine, Changchun Sci-Tech University, Changchun 130600, China
| | - Yulei Wang
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jing Zhang
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Wei Li
- College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, China.
| |
Collapse
|
32
|
Bjerring JS, Khodour Y, Peterson EA, Sachs PC, Bruno RD. Intercellular mitochondrial transfer contributes to microenvironmental redirection of cancer cell fate. FEBS J 2025; 292:2306-2322. [PMID: 39934946 PMCID: PMC12062771 DOI: 10.1111/febs.70002] [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: 05/23/2024] [Revised: 10/04/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
The mammary microenvironment has been shown to suppress tumor progression by redirecting cancer cells to adopt a normal mammary epithelial progenitor fate in vivo. However, the mechanism(s) by which this alteration occurs has yet to be defined. Here, we test the hypothesis that mitochondrial transfer from normal mammary epithelial cells to breast cancer cells plays a role in this redirection process. We evaluate mitochondrial transfer in 2D and 3D organoids using our unique 3D bioprinting system to produce chimeric organoids containing normal and cancer cells. We demonstrate that breast cancer tumoroid growth is hindered following interaction with mammary epithelial cells in both 2D and 3D environments. Furthermore, we show mitochondrial transfer occurs between donor mammary epithelial cells and recipient cancer cells primarily through tunneling nanotubes (TNTs) with minimal amounts seen from extracellular transfer of mitochondria, likely via extracellular vesicles (EVs). This organelle exchange results in various cellular and metabolic alterations within cancer cells, reducing their proliferative potential, and making them susceptible to microenvironmental control. Our results demonstrate that mitochondrial transfer contributes to microenvironmental redirection of cancer cells through alteration of metabolic and molecular functions of the recipient cancer cells. To the best of our knowledge, this is the first description of a 3D bioprinter-assisted organoid system for studying mitochondrial transfer. These studies are also the first mechanistic insights into the process of mammary microenvironmental redirection of cancer and provide a framework for new therapeutic strategies to control cancer.
Collapse
Affiliation(s)
- Julie Sofie Bjerring
- School of Medical Diagnostics and Translational Sciences, College of Health SciencesOld Dominion UniversityNorfolkVAUSA
| | - Yara Khodour
- School of Medical Diagnostics and Translational Sciences, College of Health SciencesOld Dominion UniversityNorfolkVAUSA
| | - Emilee Anne Peterson
- School of Medical Diagnostics and Translational Sciences, College of Health SciencesOld Dominion UniversityNorfolkVAUSA
| | - Patrick Christian Sachs
- School of Medical Diagnostics and Translational Sciences, College of Health SciencesOld Dominion UniversityNorfolkVAUSA
| | - Robert David Bruno
- School of Medical Diagnostics and Translational Sciences, College of Health SciencesOld Dominion UniversityNorfolkVAUSA
| |
Collapse
|
33
|
Mukherjee S, Banik SK, Chakraborty S, Das T, Choudhury MD, Tripathi A. Bryophyllum pinnatum Induces p53-Dependent Apoptosis of Colorectal Cancer Cells via Increased Intracellular ROS and G2/M Cell-Cycle Arrest In Vitro and Validated in Silico by Molecular Docking. Cell Biol Int 2025; 49:534-554. [PMID: 39992739 DOI: 10.1002/cbin.70004] [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: 09/27/2024] [Revised: 01/31/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025]
Abstract
Chemotherapy, radiotherapy and surgical treatments of cancer having several limitations and toxic side-effects, have led researchers to focus towards development of alternative natural plant-based therapeutics that can reduce disease severity. The present research work is mainly focussed towards identifying molecular mechanisms of apoptosis of colorectal cancer cells (HCT116) by perennial herb Bryophyllum pinnatum leaf-extract via both in vitro experimentations and in silico analysis. B. pinnatum leaf extract induced highest cytotoxicity at lowest dose (IC50:0.01 mg/mL) against HCT116 cells with 49.5% (p < 0.0001) cellular death, in comparison to other cancer cell lines. It has arrested HCT116 cell populations at G2/M cell-cycle phase and led to 10 folds (p < 0.0001) and 5.5 folds (p < 0.0001) increased intracellular ROS production in treated groups. ROS production might have led to significant 34.23% and 21.03% (p < 0.0001) apoptosis in treated cells, proved in vitro and in silico, with significant upregulation of p53 (p < 0.0001), BAX (p = 0.0252), CASPASE3 (p < 0.0001) and downregulation of BCL2 (p = 0.0058), leading to increased nuclear p53 (p = 0.0002) accumulation in treated cells, suggesting that the leaf-extract might have induced p53-dependent apoptosis of colorectal cancer cells. The phyto-extract also possess significant gene-modulatory potential as evident from qRT-PCR analysis of oncogenes and tumor suppressor genes. Leaf's bioactive phyto-constituents were elucidated by GC-MS and HPLC-ESI/MS analysis. In silico STITCH analysis provided significant network interactions between these bioactive phyto-compounds and studied proteins. Further Molecular Docking studies revealed strong binding between such docked complexes. Also, predicted major bioactive phyto-constituents of B. pinnatum leaf-extract such as Quercetin, Morin and β-Sitosterol have induced significant (p < 0.0001) apoptosis and increased intracellular ROS, validating their in silico interactions with studied proteins of HCT116 cells. All these studies together demonstrated ability of B. pinnatum to be used as a suitable natural phyto-therapeutic agent for development of chemo-preventive medications against colorectal cancer.
Collapse
Affiliation(s)
- Sumoyee Mukherjee
- Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical, Medicine, Kolkata, India
| | - Sheuli Kangsa Banik
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
| | | | | | | | - Anusri Tripathi
- Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical, Medicine, Kolkata, India
| |
Collapse
|
34
|
Mfotie Njoya E, Tabakam GT, Chukwuma CI, Makhafola TJ. Buxus natalensis (Oliv.) Hutch (Buxaceae) Exhibits Its Anticancer Potential by Stimulating ROS Production and Caspase-p53-BCL-2-Dependent Apoptosis in Hepatocellular Carcinoma and Prostate Cancer Cell Lines. Int J Mol Sci 2025; 26:4173. [PMID: 40362409 PMCID: PMC12071716 DOI: 10.3390/ijms26094173] [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: 03/31/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Buxus natalensis is recognized as a rich source of triterpenoidal alkaloids that are known to be effective in fighting different cancer types. Nevertheless, to date, no anticancer potential of B. natalensis extract has been yet described. Here, we investigated the antiproliferative activity of different B. natalensis leaf extracts on eight cancer cell lines (MCF-7, 4T1, Caco-2, HeLa, A549, HepG2, DU145, and LNCaP). Chang liver cell line derived from normal liver tissue, was used as control. B. natalensis hydroethanolic leaf extract (BNHLE) was found to exert significant cytotoxic effect against cancerous cell lines, with the highest efficacy being observed on LNCaP and HepG2 with IC50 values of 47.39 and 78.01 µg/mL, respectively. Interestingly, BNHLE was less cytotoxic towards Chang liver cells with an IC50 value of 334.10 µg/mL, yielding selectivity index (SI) values of 6.96 and 4.22 against LNCaP and HepG2 cells, respectively. The study of mechanism of action revealed that BNHLE exerted its antiproliferative effect by inducing ROS production and caspase -3/-7, and -9 activities in LNCaP and HepG2 cells. Moreover, it was found that BNHLE activated apoptosis in both cancerous cell lines by enhancing the expression levels of p53, while suppressing the expression of NF-κB-p65 and BCL-2 protein levels in a dose-dependent manner. The phytochemical analysis of BNHLE showed the presence of flavonoids (24.45 mgQE/g extract) and phenolics (84.64 mgGAE/g extract), and its LC-MS profiling identified several compounds including robinin and rutin, which are known for their cytotoxic effect against different cancer cell lines, such as hepatocellular carcinoma and prostate cancer cell lines. Several compounds are still unknown from B. natalensis, but the data obtained so far justify the use of B. natalensis as a potential source of bioactive compounds against hepatocellular and prostate cancers.
Collapse
Affiliation(s)
- Emmanuel Mfotie Njoya
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9301, Free State, South Africa; (G.T.T.); (C.I.C.)
| | | | | | - Tshepiso J. Makhafola
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9301, Free State, South Africa; (G.T.T.); (C.I.C.)
| |
Collapse
|
35
|
Du J, Guo B, Liu J, Li Z, Zhao X, Shao M, Yang F. Clinical pathological characteristics and prognostic analysis of renal primitive neuroectodermal tumours: a multicentre retrospective study of 16 cases in Northwest China. J Cancer Res Clin Oncol 2025; 151:151. [PMID: 40289215 PMCID: PMC12034590 DOI: 10.1007/s00432-025-06210-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025]
Abstract
OBJECTIVE Renal primitive neuroectodermal tumours (rPNETs) are extremely rare and highly aggressive malignancy, posing significant diagnostic and therapeutic challenges. This study aims to describe the clinicopathological characteristics, treatment strategies, and survival outcomes of 16 cases of rPNET from multiple centers in Northwest China, and to explore potential prognostic factors. METHODS A multicenter retrospective study was conducted, including 16 patients diagnosed with rPNET across five hospitals in Northwest China. Immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) were employed to assess the expression of molecular markers, including P53, BCL-2, Ki-67, and EWSR1 gene rearrangements. Survival analysis was performed using the Kaplan-Meier method, and prognostic factors were evaluated using univariate and multivariate Cox regression models. RESULTS The median age of the patients was 39 years, with a median Ki-67 proliferation index of 50%. P53 mutations were detected in 87.0% of cases, and BCL-2 positive expression was observed in 56.25% of cases. The median overall survival (OS) was 14 months. Univariate analysis revealed that age, tumor stage, BCL-2 expression, and Ki-67 index were significantly associated with OS. Multivariate analysis identified high Ki-67 expression (HR = 1.100, 95% CI: 1.030-1.174, p = 0.004) and negative BCL-2 expression (HR = 0.151, 95% CI: 0.026-0.888, p = 0.037) as independent risk factors for poor prognosis. Kaplan-Meier survival curves demonstrated that the median OS was significantly shorter in patients with high Ki-67 expression (12 months) compared to those with low Ki-67 expression (20 months) (Log-rank test, P < 0.01). Similarly, the median OS was significantly shorter in the BCL-2 negative group (10 months) compared to the BCL-2 positive group (24 months) (Log-rank test, P < 0.05). CONCLUSION The absence of rosette structures does not exclude the diagnosis of rPNET. BCL-2 and Ki-67 expression are significant prognostic factors, with high Ki-67 expression and negative BCL-2 expression associated with worse outcomes. These findings highlight the importance of molecular markers in risk stratification and treatment planning for rPNET.
Collapse
Affiliation(s)
- Jing Du
- Department of Pathology and Dermatology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
- Department of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Bo Guo
- Department of Burns and Plastic Surgery, Tangdu Hospital of Air Force Medical University, Xi'an, Shaanxi, 710038, China
| | - Jiayan Liu
- Xijing Hospital of Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Zhenzhen Li
- Department of Pathology and Dermatology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
| | - Xilian Zhao
- Department of Pathology, Shaanxi Provincial Tumor Hospital, Xi'an, Shaanxi, 710003, China
| | - Mingyu Shao
- Department of Pathology, Xinjiang Uygur Autonomous Region People's Hospital, Urumchi, Xinjiang, 830001, China
| | - Fan Yang
- Department of Pathology and Dermatology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China.
| |
Collapse
|
36
|
Esposito G, Ponticelli M, Milella L, Lela L, Teta R, Pawlik JR, Russo D, Costantino V. Bioassay-Guided Procedure Coupled with HR-ESIMS Dereplication for Isolation of Antiproliferative Bromo-Tyramine Derivative from Aplysina cauliformis. Mar Drugs 2025; 23:187. [PMID: 40422777 DOI: 10.3390/md23050187] [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: 03/28/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/28/2025] Open
Abstract
The marine environment is vital for sustaining life on Earth and offers a significant, untapped source of bioresources that could enhance the blue economy. The present investigation used our protocol to quickly identify bioactive molecules in Aplysina cauliformis organic extracts. This procedure combines a bioassay-guided approach with the dereplication of mass data through bioinformatic analysis. This approach identified the compound N,N,N-trimethyl-3,5-dibromotyramine, a bromo-tyramine analog that showed promising antiproliferative activity on HepG2 cell lines, with an IC50 value of 37.49 ± 1.94 μg/mL after 24 h. Furthermore, the evaluation of related gene expression confirmed the mechanism of cell death to be apoptosis. N,N,N-trimethyl-3,5-dibromotyramine increased the expression of pro-apoptotic β-cell lymphoma 2-associated X protein (BAX) and Poly (ADP-ribose) polymerase (PARP-1) cleavage (c-PARP-1) and downregulated the anti-apoptotic β-cell lymphoma 2 (BCL-2) and phospho-Akt (p-AKT). This report presents N,N,N-trimethyl-3,5-dibromotyramine from Aplysina cauliformis and its antiproliferative activity against the HepG2 cell line.
Collapse
Affiliation(s)
- Germana Esposito
- The Blue Chemistry Lab, Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Maria Ponticelli
- Department of Biochemical Pharmacology & Drug Design, Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences (BAS), Acad. G. Bonchev Str., bl. 21, 1113 Sofia, Bulgaria
- Department of Health Science, University of Basilicata, Via dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Luigi Milella
- Department of Health Science, University of Basilicata, Via dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Ludovica Lela
- Department of Health Science, University of Basilicata, Via dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Roberta Teta
- The Blue Chemistry Lab, Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Joseph R Pawlik
- Department of Biology and Marine Biology, Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| | - Daniela Russo
- Department of Health Science, University of Basilicata, Via dell'Ateneo Lucano 10, 85100 Potenza, Italy
- Spinoff BioActiPlant s.r.l., Via dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Valeria Costantino
- The Blue Chemistry Lab, Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| |
Collapse
|
37
|
Bauer JR, Robinson TL, Strich R, Cooper KF. Quitting Your Day Job in Response to Stress: Cell Survival and Cell Death Require Secondary Cytoplasmic Roles of Cyclin C and Med13. Cells 2025; 14:636. [PMID: 40358161 PMCID: PMC12071894 DOI: 10.3390/cells14090636] [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: 03/08/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Following unfavorable environmental cues, cells reprogram pathways that govern transcription, translation, and protein degradation systems. This reprogramming is essential to restore homeostasis or commit to cell death. This review focuses on the secondary roles of two nuclear transcriptional regulators, cyclin C and Med13, which play key roles in this decision process. Both proteins are members of the Mediator kinase module (MKM) of the Mediator complex, which, under normal physiological conditions, positively and negatively regulates a subset of stress response genes. However, cyclin C and Med13 translocate to the cytoplasm following cell death or cell survival cues, interacting with a host of cell death and cell survival proteins, respectively. In the cytoplasm, cyclin C is required for stress-induced mitochondrial hyperfission and promotes regulated cell death pathways. Cytoplasmic Med13 stimulates the stress-induced assembly of processing bodies (P-bodies) and is required for the autophagic degradation of a subset of P-body assembly factors by cargo hitchhiking autophagy. This review focuses on these secondary, a.k.a. "night jobs" of cyclin C and Med13, outlining the importance of these secondary functions in maintaining cellular homeostasis following stress.
Collapse
Affiliation(s)
| | | | | | - Katrina F. Cooper
- Department of Cell and Molecular Biology, School of Osteopathic Medicine, Rowan-Virtua College of Medicine and Life Sciences, Rowan University, Stratford, NJ 08084, USA; (J.R.B.); (T.L.R.); (R.S.)
| |
Collapse
|
38
|
Hafez MH, El-Far AH, Elblehi SS. Astaxanthin alleviates fipronil-induced neuronal damages in male rats through modulating oxidative stress, apoptosis, and inflammatory markers. Sci Rep 2025; 15:14299. [PMID: 40274901 PMCID: PMC12022106 DOI: 10.1038/s41598-025-95447-3] [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: 01/03/2025] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
Fipronil (FPN) is an effective pesticide for veterinary and agricultural use; however, it can induce neurotoxic effects on non-target organisms after accidental exposure. Astaxanthin (AST) is a dark red carotenoid with antioxidant, anti-inflammatory, neuroprotective, and antiapoptotic effects. This study investigated the ameliorative impact of AST against FPN-induced brain damage in rats. Thirty-two adult Wistar rats were allocated into four groups (n = 8): Control, AST (20 mg/kg bwt/day), fipronil (FPN) (20 mg/kg bwt/day), and AST + FPN group. Acetylcholine (ACh), dopamine, malondialdehyde (MDA), and proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and inflammatory cytokine cyclooxygenase-2 (COX2) levels were enhanced in the FPN-administered group relative to the control group. In addition, a substantial reduction of acetylcholine esterase (AchE), gamma-aminobutyric acid (GABA), serotonin, reduced glutathione (GSH) levels, catalase (CAT), and total superoxide dismutase (T-SOD) enzyme activities were determined. FPN induced histopathological alterations in the cerebral and cerebellar tissues. Likewise, the histomorphometric image analysis of H and E-stained tissue sections was constant with FPN-induced neurotoxicity. Immunohistochemically, an intense positive immunohistochemical staining of apoptotic marker caspase-3 and astrocytes activation marker glial fibrillary acidic protein (GFAP) in the examined tissues was noticed. Inversely, the simultaneous administration of AST partially attenuated FPN impacts, ameliorating the severity of FPN-induced neuronal damage. These results were also established with the molecular docking findings. It could be suggested that AST has antioxidant, anti-inflammatory, and anti-apoptotic capabilities against FPN-induced neuronal damage via suppression of oxidative stress and pro-inflammatory cytokines, preservation of the neurotransmitters, and the cerebral and cerebellar histoarchitectures.
Collapse
Affiliation(s)
- Mona H Hafez
- Department of Physiology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22758, Egypt.
| | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22758, Egypt
| |
Collapse
|
39
|
Cai JL, Zhang Y, Gao H, Wang Q, Huang W, Cai YJ, Jia WX, Wang JJ, Chen X, Sun HY. Molecular characterization, expression pattern and the function of TRAF2 from blood parrot Amphilophus citrinellus ×Vieja melanura response to LPS stimulation. FISH & SHELLFISH IMMUNOLOGY 2025; 163:110362. [PMID: 40280260 DOI: 10.1016/j.fsi.2025.110362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
Tumor necrosis factor receptor-associated factor (TRAF) family is a critical signal transduction protein, and plays important roles in cell growth, apoptosis, and immune response, etc. In this study, molecular characteristics, expression patterns, and the role of TRAF2 in blood parrot Vieja synspila ♀ × Amphilophus citrinellus ♂, an important ornamental fish, were explored response to lipopolysaccharide (LPS) challenge. The full length of blood parrot TRAF2 was 2725 bp, with an open reading frame (ORF) of 1551 bp encoding 516 amino acids, and a molecular weight of 58.58 kDa. Blood parrot TRAF2 contained four conserved domains: RING, TRAF-type zinc finger, TRAF_BIRC3_bd, and MATH (Meprin and TRAF-C homology). Analysis of phylogenetic relationships showed that TRAF2 were conserved in different species, indicating that its role might be similar. Blood parrot TRAF2 mRNA could be detected in all of the tissues examined, and was distributed in both the cytoplasm and nucleus. The expression of blood parrot TRAF2 was up-regulated during LPS challenge. Overexpression of TRAF2 could significantly inhibit the activities of nuclear factor κB (NF-κB) and activated protein 1 (AP-1), and reduce the ratio of Bax/Bcl-2. This study indicated that the TRAF2 might play important roles in organisms during pathogen infection.
Collapse
Affiliation(s)
- Jie-Li Cai
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; School of Life Sciences, South China Normal University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yue Zhang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Hui Gao
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Qi Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wei Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yi-Jie Cai
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wei-Xin Jia
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jun-Jie Wang
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong, China.
| | - Xiao Chen
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.
| | - Hong-Yan Sun
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China.
| |
Collapse
|
40
|
Liu Z, Lenz HJ, Yu J, Zhang L. Differential Response and Resistance to KRAS-Targeted Therapy. Mol Carcinog 2025. [PMID: 40256920 DOI: 10.1002/mc.23908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/10/2025] [Indexed: 04/22/2025]
Abstract
KRAS is the most frequently mutated oncogene. In epithelial malignancies such as lung, colorectal, and pancreatic tumors, KRAS is mutated in 25 to above 90% cases. KRAS was considered undruggable for over three decades until the recent development of covalent inhibitors targeting the KRAS G12C mutant. The recent approval of the KRAS G12C inhibitors sotorasib and adagrasib has ushered in a new era of KRAS-targeted therapy. Despite this success, a major challenge in KRAS-targeted therapy is intrinsic and acquired resistance to KRAS inhibitors. Clinical studies have shown that many patients with KRAS G12C cancers did not respond to sotorasib and adagrasib. Colorectal cancer, in particular, has a markedly lower response rate to KRAS G12C inhibitors compared to non-small cell lung cancer. Furthermore, the therapeutic response to KRAS G12C inhibition was short-lived, with quick emergence of acquired resistance. In this review, we summarize several major themes that have emerged from recent clinical and preclinical studies on the mechanisms of intrinsic and acquired resistance to KRAS-targeted therapy in colorectal, lung, and pancreatic cancers. We also discuss various combination strategies for targeting these mechanisms to overcome resistance to KRAS inhibitors.
Collapse
Affiliation(s)
- Zhaojin Liu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Heinz-Josef Lenz
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Jian Yu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Lin Zhang
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| |
Collapse
|
41
|
Duzgun D, Oltean S. Aberrant Splicing as a Mechanism for Resistance to Cancer Therapies. Cancers (Basel) 2025; 17:1381. [PMID: 40282556 PMCID: PMC12025770 DOI: 10.3390/cancers17081381] [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: 02/17/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Cancer is biologically diverse, highly heterogeneous, and associated with molecular alterations, significantly contributing to mortality worldwide. Currently, cancer patients are subjected to single or combination treatments comprising chemotherapy, surgery, immunotherapy, radiation therapy, and targeted therapy. Chemotherapy remains the first line of treatment in cancer but faces a major obstacle in the form of chemoresistance. This obstacle has resulted in relapses and poor patient survival due to decreased treatment efficacy. Aberrant pre-mRNA alternative splicing can significantly modulate gene expression and function involved in the resistance mechanisms, potentially shaping the intricate landscape of tumour chemoresistance. Thus, novel strategies targeting abnormal pre-mRNA alternative splicing and understanding the molecular mechanisms of chemotherapy resistance could aid in overcoming the chemotherapeutic challenges. This review first highlights drug targets, drug pumps, detoxification mechanisms, DNA damage response, and evasion of apoptosis and cell death as key molecular mechanisms involved in chemotherapy resistance. Furthermore, the review discusses the progress of research on the dysregulation of alternative splicing and molecular targets involved in chemotherapy resistance in major cancer types.
Collapse
Affiliation(s)
| | - Sebastian Oltean
- Department of Clinical and Biomedical Sciences, Faculty of Health Sciences, University of Exeter, Exeter EX1 2LU, UK
| |
Collapse
|
42
|
Ozerklig B, Turkel I, Yilmaz M, Vaizoglu RD, Akan HS, Dikmen ZG, Saleem A, Kosar SN. Exercise-induced extracellular vesicles mediate apoptosis in human colon cancer cells in an exercise intensity-dependent manner. Eur J Appl Physiol 2025:10.1007/s00421-025-05787-1. [PMID: 40253655 DOI: 10.1007/s00421-025-05787-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/26/2025] [Indexed: 04/22/2025]
Abstract
Regular exercise reduces the incidence and improves the prognosis of many cancer types, but the underlying mechanisms remain elusive. Evidence suggests that exercise exerts its therapeutic effects through extracellular vesicles (EVs), which are essential for cellular communication. Here, we hypothesized that exercise-induced EVs from serum of healthy individuals would exert anti-tumorigenic effects on human colon cancer HT-29 cells, in an exercise intensity-dependent manner. Ten healthy young active males participated in a randomized crossover trial, completing two workload-matched acute exercise bouts, moderate-intensity continuous exercise (MICE) and high-intensity interval exercise (HIIE), on a cycle ergometer. A control session of rest (PRE) was included. EVs were isolated from serum samples collected during PRE and immediately after each exercise session. EVs were co-incubated with HT-29 colon cancer cells, and the effects on cell viability, migration, and apoptosis were measured. EV treatment reduced cell viability in all groups (PRE, MICE, and HIIE) by 35%, 43% and 47%, respectively, vs. PBS. HIIE-EVs showed a significantly greater reduction in cell viability vs. PRE; therefore, only these groups were used for further analysis. PRE EVs reduced migration by 27%, and HIIE-EVs by 39%. HIIE-EVs increased expression of pro-apoptotic markers: Bax/Bcl-2 ratio by 56% and Caspase 3 by 30% vs. PBS, with no change observed in the PRE group. Further, 16% of cells in PRE and 28% of cells in HIIE were TUNEL-positive, indicating DNA fragmentation. To our knowledge, this is the first human study that illustrates the therapeutic potential of exercise-induced EVs in cancer treatment.
Collapse
Affiliation(s)
- Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye.
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada.
- The Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, Canada.
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| | - Merve Yilmaz
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Refika Dilara Vaizoglu
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Türkiye
| | - Handan Sevim Akan
- Department of Biology, Faculty of Science, Hacettepe University, Ankara, Türkiye
| | - Z Gunnur Dikmen
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Ayesha Saleem
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada
- The Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, Canada
| | - Sukran Nazan Kosar
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
43
|
Singh M, Arora HL, Naik R, Joshi S, Sonawane K, Sharma NK, Sinha BK. Ferroptosis in Cancer: Mechanism and Therapeutic Potential. Int J Mol Sci 2025; 26:3852. [PMID: 40332483 PMCID: PMC12028135 DOI: 10.3390/ijms26083852] [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: 02/11/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer drug resistance occurs when cancer cells evade cell death following treatment with chemotherapy, radiation therapy, and targeted therapies. This resistance is often linked to the reprogramming of programmed cell death (PCD) pathways, allowing cancer cells to survive drug-induced stress. However, certain anticancer therapies, when combined with specific agents or inhibitors, can induce ferroptosis-a form of cell death driven by iron-dependent lipid peroxidation. Currently, extensive preclinical and clinical research is underway to investigate the molecular, cellular, and tissue-specific mechanisms underlying ferroptosis, with the goal of identifying strategies to overcome drug resistance in cancers unresponsive to conventional PCD pathways. By harnessing ferroptosis, cancer cells can be compelled to undergo lipid peroxidation-induced death, potentially improving therapeutic outcomes in patients with cancer. This short review aims to enhance the understanding of ferroptosis inducers in cancer therapy and stimulate further research into ferroptosis-based approaches for more effective clinical cancer treatment.
Collapse
Affiliation(s)
- Mansaa Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Hasmiq L. Arora
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Rutuja Naik
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Shravani Joshi
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Kaveri Sonawane
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Birandra K. Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
44
|
Ahmed HS. Neuropharmacological effects of calycosin: a translational review of molecular mechanisms and therapeutic applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04154-3. [PMID: 40237798 DOI: 10.1007/s00210-025-04154-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Calycosin, a naturally occurring isoflavonoid found predominantly in Astragalus membranaceus, exhibits significant therapeutic potential in various neurological conditions. Its multifaceted bioactive properties-antioxidant, anti-inflammatory, and anti-apoptotic-position it as a promising candidate for neuroprotection and neuroregeneration. This review explores calycosin's mechanisms of action, including its modulation of key signaling pathways such as HMGB1/TLR4/NF-κB (high mobility group box 1/toll-like receptor 4/nuclear factor kappa B), phosphatidylinositol-3-kinase (PI3 K)/Akt, ERK1/2 (extracellular signal-regulated kinase 1/2), and Hsp90/Akt/p38. In cerebral ischemia/reperfusion injury, calycosin reduces oxidative stress markers like ROS (reactive oxygen species) and MDA (malondialdehyde), enhances antioxidant enzymes (SOD (superoxide dismutase) and GPX (glutathione peroxidase)), and downregulates pro-inflammatory cytokines (TNF-α, IL-1β) through the HMGB1/TLR4/NF-κB pathway. It also inhibits autophagy via the STAT3/FOXO3a pathway and apoptosis by modulating Bax and Bcl-2 expression. In neuro-oncology, calycosin inhibits glioblastoma cell migration and invasion by modulating the TGF-β-mediated mesenchymal properties and suppressing the c-Met and CXCL10 signaling pathways. Additionally, it enhances the efficacy of temozolomide in glioma treatment through apoptotic pathways involving caspase-3 and caspase-9. Calycosin shows promise in Alzheimer's disease by reducing β-amyloid production and tau hyperphosphorylation via the GSK-3β pathway and improving mitochondrial function through the peroxisome proliferator-activated receptor gamma coactivator 1-Alpha (PGC-1α)/mitochondrial transcription factor A (TFAM) signaling pathway. In Parkinson's disease, calycosin mitigates oxidative stress, prevents dopaminergic neuronal death, and reduces neuroinflammation by inhibiting the TLR/NF-κB and MAPK pathways. It has also shown therapeutic potential in meningitis and even neuroprotective effects against hyperbilirubinemia-induced nerve injury. Despite these promising findings, further research, including detailed mechanistic studies and clinical trials, is needed to fully understand calycosin's therapeutic mechanisms and validate its potential in human subjects. Developing advanced delivery systems and exploring synergistic therapeutic strategies could further enhance its clinical application and effectiveness.
Collapse
Affiliation(s)
- H Shafeeq Ahmed
- Bangalore Medical College and Research Institute, K.R Road, Bangalore, 560002, Karnataka, India.
| |
Collapse
|
45
|
Al-Wahaibi LH, Elshamsy AM, Ali TFS, Youssif BGM, Bräse S, Abdel-Aziz M, El-Koussi NA. Design, synthesis, in silico studies, and apoptotic antiproliferative activity of novel thiazole-2-acetamide derivatives as tubulin polymerization inhibitors. Front Chem 2025; 13:1565699. [PMID: 40308265 PMCID: PMC12040969 DOI: 10.3389/fchem.2025.1565699] [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: 01/23/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction Tubulin polymerization inhibitors have emerged as interesting anticancer therapies. We present the design, synthesis, and structural elucidation of novel thiazole-based derivatives to identify novel tubulin inhibitors with potent antiproliferative efficacy and strong inhibition of tubulin polymerization. Methods The novel compounds consist of two scaffolds. Scaffold A compounds 10a-e and scaffold B compounds 13a-e. the structures of the newly synthesized compounds 10a-e and 13a-e were validated using 1H NMR, 13C NMR, and elemental analysis. Results and Discussion The most effective antitubulin derivative was 10a, exhibiting an IC50 value of 2.69 μM. Subsequently, 10o and 13d exhibited IC50 values of 3.62 μM and 3.68 μM, respectively. These compounds exhibited more potency than the reference combretastatin A-4, which displayed an IC50 value of 8.33 μM. These compounds had no cytotoxic effects on normal cells, preserving over 85% cell viability at 50 μM. The antiproliferative experiment demonstrated that compounds 10a, 10o, and 13d displayed significant activity against four cancer cell lines, with average GI50 values of 6, 7, and 8 μM, equivalent to the reference's doxorubicin and sorafenib. Compounds 10a, 10o, and 13d were demonstrated to activate caspases 3, 9, and Bax, while down-regulating the anti-apoptotic protein Bcl2. Molecular docking studies demonstrated superior binding affinities for 10a (-7.3 kcal/mol) at the colchicine binding site of tubulin, forming key hydrophobic and hydrogen bonding interactions that enhance its activity. ADMET analysis confirmed favorable drug-like properties, establishing these compounds as promising candidates for further development as anticancer agents targeting tubulin polymerization.
Collapse
Affiliation(s)
- Lamya H. Al-Wahaibi
- Department of Chemistry, College of Sciences, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ali M. Elshamsy
- Pharmceutical Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Taha F. S. Ali
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Bahaa G. M. Youssif
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Nawal A. El-Koussi
- Pharmceutical Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
46
|
Saadh MJ, Muhammad FA, Alazzawi TS, Fahdil AA, Athab ZH, Tuxtayev J, Alsaikhan F, Farhood B. Regulation of Apoptotic Pathways by MicroRNAs: A Therapeutic Strategy for Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04833-5. [PMID: 40220245 DOI: 10.1007/s12035-025-04833-5] [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: 12/06/2024] [Accepted: 03/09/2025] [Indexed: 04/14/2025]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder marked by a gradual decline in memory and cognitive functions. It is characterized by the presence of senile plaques, neurofibrillary tangles, and neuronal degeneration, affecting a significant portion of the human population. A key feature of various nervous system disorders, including AD, is extensive cellular death caused by apoptosis, which affects not only neurons but also glial cells. While apoptosis plays a vital role in eliminating certain cells and supporting normal development, alterations or disruptions in apoptotic pathways can lead to harmful neurodegenerative conditions such as AD. Thus, targeting apoptosis presents a promising therapeutic approach for these diseases. MicroRNAs (miRNAs), a class of non-coding RNA, play diverse roles in cellular functions, including proliferation, gene expression regulation, programmed cell death, intercellular communication, and angiogenesis. By modulating regulatory genes, miRNAs can influence apoptosis, either promoting or inhibiting it. Aberrant expression of miRNAs can impact multiple apoptotic pathways, potentially driving the progression of AD and related health issues. This review summarizes recent research on miRNAs and their dual role in exacerbating or protecting against neural cell damage in AD by altering apoptotic pathways. The regulation of apoptosis by miRNAs offers a prospective therapeutic strategy for Alzheimer's disease.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Tuqa S Alazzawi
- College of Dentist, National University of Science and Technology, Dhi Qar, Iraq
| | - Ali A Fahdil
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Jamshid Tuxtayev
- Department of Surgical Diseases, Faculty of Pediatrics, Samarkand State Medical Institute, Samarkand, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
47
|
White TLA, Henry DJ, Roberts SDA, Jin Y, Gong YN, Morel PA. HnRNP L is essential for peripheral T cell proliferation and survival. Front Immunol 2025; 16:1543145. [PMID: 40276511 PMCID: PMC12018431 DOI: 10.3389/fimmu.2025.1543145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/14/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction During T cell development, heterogeneous nuclear ribonucleoprotein (hnRNP) L is known to regulate CD4 T helper subset differentiation, the proliferation and migration of thymocytes, as loss of hnRNP L in early T cell development results in a failure of T cells to reach the periphery. Methods To better understand the role of hnRNP L in modulating peripheral T cell trafficking and function, we analyzed T survival and activation in newly generated CD4Cre x hnRNP LFl/Fl (KO) mice. In vitro and in vivo analyses of CD4 T cell differentiation, T cell proliferation and death post activation were performed. Results Our initial study of the steady state profile of the KO mice showed normal migration of T cells from the thymus, but peripheral T cell numbers were reduced. Analysis of TCR-mediated signaling pathways revealed normal early T cell activation. However, T cells lacking hnRNP L had marked defects in their ability to differentiate into T helper cell subsets due to reduced proliferation and increased death. In vivo, using immunization studies, KO CD4 T cells failed to fully differentiate into T follicular helper (Tfh) cells and were unable to support the formation of germinal center B cells. Death of activated hnRNP L KO cells could be reversed by treating the cells with zVAD, a pan-caspase inhibitor. In addition, hnRNP L KO cells failed to upregulate the anti-apoptotic protein Bcl-XL following activation. Discussion These studies suggest that hnRNP L plays an important role in T cell activation and survival. Our studies suggest that hnRNP L plays a critical pro-survival role in activated T cells and that alternative splicing of factors that prevent apoptosis may be an important mechanism by which this is achieved.
Collapse
Affiliation(s)
| | | | | | | | | | - Penelope A. Morel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
48
|
Panda PK, Paschoalini Mafra AC, Bastos AC, Cao L, Serra Bonet M, Brashears CB, Chen EY, Benedict-Hamilton HM, Ehrhardt W, Bomalaski J, Dehner C, Rogers LC, Oyama T, Van Tine BA. BCL-XL Protects ASS1-Deficient Cancers from Arginine Starvation-Induced Apoptosis. Clin Cancer Res 2025; 31:1333-1345. [PMID: 39898973 PMCID: PMC11964295 DOI: 10.1158/1078-0432.ccr-24-2548] [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: 08/06/2024] [Revised: 12/26/2024] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE Argininosuccinate synthetase 1 (ASS1) silencing in carcinomas and sarcomas leads to a dependence on extracellular arginine for survival. Arginine deprivation therapies, such as PEGylated arginine deiminase (ADI-PEG20), have shown limited effectiveness, which may be due to underlying mechanisms that inhibit apoptosis. EXPERIMENTAL DESIGN The effects of ADI-PEG20 on cell-cycle regulation, apoptosis, and BCL-XL-mediated survival pathways in ASS1-deficient cancer cells were determined. The mechanism of cell death protection was determined by assessing caspase and PARP cleavage, CDK2 activity, MCL1 expression, and the interactions among BCL-XL, BAX, and BAK. In vitro synergy was determined, and in vivo efficacy was modeled. RESULTS Treatment with ADI-PEG20 led to reduced CDK2 activity and inhibited cell-cycle progression but did not induce significant cell death. BCL-XL was found to bind to BAX and BAK, preventing the initiation of apoptosis despite arginine starvation. Inhibition of BCL-XL allowed proapoptotic BAX and BAK to initiate the intrinsic apoptosis pathway, leading to increased cell death. This was found to be synergistic in vitro and efficacious in combination in vivo. CONCLUSIONS The study identifies BCL-XL as a key factor limiting the efficacy of arginine starvation therapies. Combining BCL-XL inhibitors with arginine deprivation strategies may overcome this resistance and enhance therapeutic outcomes. These findings provide a strong preclinical rationale for testing this combination approach in phase 1 clinical trials for ASS1-deficient cancers.
Collapse
Affiliation(s)
- Prashanta Kumar Panda
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Ana Carolina Paschoalini Mafra
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Alliny C.S. Bastos
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Li Cao
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Maria Serra Bonet
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Caitlyn B. Brashears
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Ethan Yang Chen
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Heather M. Benedict-Hamilton
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - William Ehrhardt
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | | | - Carina Dehner
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Leonard C. Rogers
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Toshinao Oyama
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Brian A. Van Tine
- Division of Medical Oncology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
- Siteman Cancer Center, St. Louis, Missouri
- Department of Pediatric Hematology/Oncology, St. Louis Children’s Hospital, St. Louis, Missouri
| |
Collapse
|
49
|
Tkachenko A, Havranek O. Cell death signaling in human erythron: erythrocytes lose the complexity of cell death machinery upon maturation. Apoptosis 2025; 30:652-673. [PMID: 39924584 PMCID: PMC11947060 DOI: 10.1007/s10495-025-02081-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/11/2025]
Abstract
Over the recent years, our understanding of the cell death machinery of mature erythrocytes has been greatly expanded. It resulted in the discovery of several regulated cell death (RCD) pathways in red blood cells. Apoptosis (eryptosis) and necroptosis of erythrocytes share certain features with their counterparts in nucleated cells, but they are also critically different in particular details. In this review article, we summarize the cell death subroutines in the erythroid precursors (apoptosis, necroptosis, and ferroptosis) in comparison to mature erythrocytes (eryptosis and erythronecroptosis) to highlight the consequences of organelle clearance and associated loss of multiple components of the cell death machinery upon erythrocyte maturation. Recent advances in understanding the role of erythrocyte RCDs in health and disease have expanded potential clinical applications of these lethal subroutines, emphasizing their contribution to the development of anemia, microthrombosis, and endothelial dysfunction, as well as their role as diagnostic biomarkers and markers of erythrocyte storage-induced lesions. Fas signaling and the functional caspase-8/caspase-3 system are not indispensable for eryptosis, but might be retained in mature erythrocytes to mediate the crosstalk between both erythrocyte-associated RCDs. The ability of erythrocytes to switch between eryptosis and necroptosis suggests that their cell death is not a simple unregulated mechanical disintegration, but a tightly controlled process. This allows investigation of eventual pharmacological interventions aimed at individual cell death subroutines of erythrocytes.
Collapse
Affiliation(s)
- Anton Tkachenko
- First Faculty of Medicine, BIOCEV, Charles University, Průmyslová 595, 25250, Vestec, Czech Republic.
| | - Ondrej Havranek
- First Faculty of Medicine, BIOCEV, Charles University, Průmyslová 595, 25250, Vestec, Czech Republic
- First Department of Medicine - Hematology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
50
|
Rafiee J, Jamialahmadi K, Bazyari MJ, Aghaee-Bakhtiari SH. Drug repositioning in castration-resistant prostate cancer using systems biology and computational drug design techniques. Comput Biol Chem 2025; 115:108329. [PMID: 39731827 DOI: 10.1016/j.compbiolchem.2024.108329] [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: 04/06/2024] [Revised: 05/07/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND AND OBJECTIVE Castration-resistant prostate cancer (CRPC) is caused by resistance to androgen deprivation treatment and leads to the death of patients and there is almost no chance of survival. Therefore, finding a cure to overcome CRPC is challenging and important, but discovering a new drug is very time-consuming and expensive. To overcome these problems, we used Drug repositioning (drug repurposing) strategy in this study. METHODS Gene expression data of CRPC and primary prostate samples were extracted from the GEO database to identify DEGs. Pathway enrichment was performed to find the role of DEGs in signaling pathways. To identify hub proteins, the PPI network was reconstructed and analyzed. drug candidates were identified and to select the most effective drug, molecular docking analysis, and molecular dynamics simulation were performed. Then MTT and qRT-PCR tests were performed to check the effectiveness of the selected drug. RESULTS A total of 152 upregulated DEGs and 343 downregulated DEGs were identified, and after PPI network analysis, IKBKB, SNAP23, MYC, and NOTCH1 genes were introduced as hubs. drug candidates for IKBKB were identified and by examining the results of docking screening and molecular dynamics, sulfasalazine was selected as the most effective drug. Laboratory analyses proved the effectiveness of this drug and a decrease in the expression of all target genes was observed. CONCLUSION In this study, IKBKB key protein were identified in CRPC, and sulfasalazine was selected as a suitable candidate for drug repositioning and its effectiveness was confirmed through tests.
Collapse
Affiliation(s)
- Javad Rafiee
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Javad Bazyari
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|