1
|
Atlasbaf A, Kodehei AH, Bahadoran E, Zare I, Yousefzadeh S, Kesharwani P, Naderi Y, Sahebkar A. Bavachinin attenuates cerebral ischemia/reperfusion injury in rats via its anti-inflammatory and antioxidant effects. Tissue Cell 2025; 95:102886. [PMID: 40164017 DOI: 10.1016/j.tice.2025.102886] [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: 12/01/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025]
Abstract
Cerebral ischemia is associated with memory deficits. Bavachinin, a natural flavonoid derived from Psoralea Corylifolia seeds, exhibits various pharmacological properties, including anti-inflammatory, antioxidant, anticancer, and anti-allergic activities. We looked into the neuroprotective effects of bavachinin on rats' memory impairments brought on by transient cerebral ischemia/reperfusion. Blocked carotid arteries caused transient cerebral ischemia/reperfusion injury. Wistar male rats were randomized to bavachinin, ischemia/reperfusion, and sham groups. After surgery, bavachinin (100 mg/kg) was injected intraperitoneally once a day for seven days. Spatial memory was evaluated using the Morris water maze test. The vitality of the hippocampal pyramidal neurons was assessed by Nissl staining. The production of pro-inflammatory cytokines (TNF-α and IL-1β) has been detected using ELISA. Oxidative stress was evaluated by determining the hippocampal levels of malondialdehyde (MDA). In rats with transient cerebral ischemia/reperfusion, bevacichinin markedly improved the performance of learning. The bavachinin-treated group had a higher number of surviving pyramidal neurons, as demonstrated by the Nissl staining. In the hippocampus, bevacichinin lowered MDA, TNF-α, and IL-1β levels. The antioxidant and anti-inflammatory properties of bavachinin likely protect against memory impairment caused by transient cerebral ischemia. These findings suggest that bavachinin could be a potent therapeutic agent for the prevention of cognitive deficits and neuronal damage associated with cerebral ischemia/reperfusion. More studies are needed to explore its clinical applications and mechanisms of action.
Collapse
Affiliation(s)
| | | | - Ensiyeh Bahadoran
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Isareza Zare
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Salar Yousefzadeh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh, India; University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Yazdan Naderi
- Cellular and Molecular Research Center, Research Institute for Prevention of Noncommunicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Thamizhmaran R, Kalaimurugan G, Siddiqui MK, Vinnarasi L, Yuvaraj A, Hanif MF. Utilizing neighborhood topological indices for QSPR analysis of clinically approved immunosuppressive drugs in heart transplant therapy. Comput Biol Chem 2025; 117:108414. [PMID: 40086343 DOI: 10.1016/j.compbiolchem.2025.108414] [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: 01/16/2025] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
Heart transplantation is a life-saving transplantation procedure for individuals with advanced heart failure who have gone through all other medicinal options. It is predicted that 5000 heart transplants will be performed annually worldwide. The immunosuppressive drugs are used after a heart transplant to prevent organ rejection. They may be administered both before and throughout the transplant process under specific circumstances. Quantitative Structure-Activity or Property Relationship using topological descriptors is essential in drug design since it allows one to anticipate the physicochemical characteristics of medications based on their molecular structure. This study investigates the neighborhood topological descriptors of immunosuppressive medications used to treat heart transplant patients. The highest predictive efficacy of the pharmaceuticals is demonstrated by the good association between the topological indicators and the physical characteristics of the transplant medications. Additionally, this data may be used by researchers to develop new and effective medications for recipients of heart transplants.
Collapse
Affiliation(s)
- R Thamizhmaran
- Department of Mathematics, Thiruvalluvar University, Vellore, 632 115, Tamil Nadu, India.
| | - G Kalaimurugan
- Department of Mathematics, Thiruvalluvar University, Vellore, 632 115, Tamil Nadu, India.
| | - Muhammad Kamran Siddiqui
- Department of Mathematics, COMSATS University Islamabad, Lahore Campus, 54000, Lahore, Pakistan.
| | - L Vinnarasi
- Department of Mathematics, Thiruvalluvar University, Vellore, 632 115, Tamil Nadu, India.
| | - A Yuvaraj
- Department of Mathematics, Thiruvalluvar University, Vellore, 632 115, Tamil Nadu, India.
| | - Muhammad Faisal Hanif
- Department of Mathematics and Statistics, The University of Lahore, Lahore Campus, 54000, Lahore, Pakistan.
| |
Collapse
|
3
|
Arghidash F, Gheybi F, Gholamhosseinian H, Kesharwani P, Sahebkar A. Radioprotective and radiosensitizing properties of silymarin/silibinin in response to ionizing radiation. Pathol Res Pract 2025; 270:156002. [PMID: 40347920 DOI: 10.1016/j.prp.2025.156002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 05/06/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Cancer is a health and treatment challenge that the world is facing, and many efforts are being made to develop treatment solutions for all forms of cancer. Radiotherapy (RT), one of the cancer treatment methods, can cause toxicity in healthy cells, even though it has positive effects on killing cancer cells. It is possible for cancer cells to develop resistance to radiotherapy. To address these issues, it can be beneficial to combine treatments. Combining plants with conventional cancer treatment is a viable option, and their potential can be utilized in this area. The therapeutic properties of silymarin and its active ingredient silibinin have been used in traditional medicine for a long time. The purpose of this review is to investigate the radioprotective and radio-sensitizing properties of silymarin/silibinin in cancer treatment.
Collapse
Affiliation(s)
- Faezeh Arghidash
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Gheybi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India; University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Cheng X, Hemmati S, Pirhayati M, Zangeneh MM, Veisi H. Decoration of copper nanoparticles (Cu 2O NPs) over chitosan-guar gum: Its application in the Sonogashira cross-coupling reactions and treatment of human lung adenocarcinoma. Int J Biol Macromol 2025; 305:141122. [PMID: 39965696 DOI: 10.1016/j.ijbiomac.2025.141122] [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: 12/01/2024] [Revised: 02/02/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
This study outlines the sustainable synthesis of hybrid biopolymer hydrogels supported with octahedral Cu2O nanoparticles (NPs), alongside their biological assessments and characterizations. A composite hydrogel made of chitosan and guar gum (CS-GG) was employed as a template for the environmentally friendly synthesis of nanoparticles. Leveraging their electron-rich functional groups, the biopolymers acted as stabilizing agents for the Cu2O NPs and as green reductants, facilitating the reduction of copper ions. The material's physicochemical properties were thoroughly examined using advanced techniques, such as X-ray diffraction (XRD), Field-Emission Scanning Electron Microscopes (FE-SEM), Eneregy Dispersive X-ray Electron Spectroscopy (EDX), Fourier Transformed Infrared Spectroscopy (FT-IR), Transmission Electron Microscopy (TEM) and ICP-OES. The resulting CS-GG/Cu2O NPs nanocomposite was investigated as a reusable heterogeneous nanocatalyst, demonstrating its efficiency in the phosphine-free, palladium-free, and ligand-free synthesis of various stilbene derivatives with high yields through the Sonogashira coupling reaction. The catalyst showed no significant reduction in activity after being reused seven times consecutively. The cytotoxic effects of the CS-GG/Cu2O NPs nanocomposite on NCI-H661 lung cancer cells and normal cells (HUVEC) were assessed over 48 h using MTT assay. The cancer cell's viability decreased after exposure to the CS-GG/Cu2O NPs, with an IC50 value of 82 μg/mL. The CS-GG/Cu2O NPs nanocomposite controls the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) system, which in turn impacts apoptosis and cell proliferation in NCI-H661 cells, according to a detailed examination of the mTOR pathway. The pathway could act a role in the cell cycle inhibition and apoptosis induced by the CS-GG/Cu2O NPs nanocomposite. The CS-GG/Cu2O NPs nanocomposite could be a useful natural anti-cancer agent for the treatment of lung cancer.
Collapse
Affiliation(s)
- Xiongtao Cheng
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Saba Hemmati
- Department of Chemistry, Payame Noor University, Tehran, Iran
| | - Mozhgan Pirhayati
- Department of Applied Chemistry, Faculty of Science, Malayer University, Malayer, Iran.
| | - Mohamad Mehdi Zangeneh
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Hojat Veisi
- Department of Chemistry, Payame Noor University, Tehran, Iran.
| |
Collapse
|
5
|
Borjian N, Farhadian S, Shareghi B, Asgharzadeh S, Momeni L, Ghobadi S. Binding affinity and mechanism of dicofol-lysozyme interaction: Insights from multi-spectroscopy and molecular dynamic simulations. Int J Biol Macromol 2025; 308:142569. [PMID: 40157692 DOI: 10.1016/j.ijbiomac.2025.142569] [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: 08/31/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
The pervasive use of dicofol in agricultural settings has been linked to biomolecular perturbations, posing significant threats to environmental sustainability and human health. Therefore, it is crucial to investigate the interactions between dicofol and biomacromolecules, such as proteins. This study employed a range of molecular modeling approaches and spectroscopic techniques to examine the binding interaction between dicofol and lysozyme to elucidate the underlying mechanisms of these toxic effects. Molecular docking studies identified the most optimal binding site for dicofol on the lysozyme structure, highlighting the precise region within the protein where dicofol binds most effectively. Molecular dynamic simulations showed that the dicofol-lysozyme system was stable throughout the entire simulation period. UV-vis absorption and fluorescence emission studies confirmed that dicofol interacts with lysozyme to form a complex. FT-IR analysis revealed that this interaction alters lysozyme's conformation, decreasing alpha-helical content while increasing β-sheet content. Furthermore, a direct relationship was observed between dicofol concentration and lysozyme's activity and stability, with higher dicofol levels causing a notable decline in both factors. In conclusion, this research deepens our understanding of the specific interactions between dicofol and lysozyme while also highlighting the importance of studying such interactions to evaluate the environmental and health risks linked to pesticide usage.
Collapse
Affiliation(s)
- Negar Borjian
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, P. O. Box.115, Iran
| | - Sadegh Farhadian
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, P. O. Box.115, Iran; Central Laboratory, Shahrekord University, Shahrekord, Iran.
| | - Behzad Shareghi
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, P. O. Box.115, Iran; Central Laboratory, Shahrekord University, Shahrekord, Iran
| | - Sanaz Asgharzadeh
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, P. O. Box.115, Iran; Central Laboratory, Shahrekord University, Shahrekord, Iran
| | - Lida Momeni
- Department of Biology, Faculty of Science, University of Payam Noor, Iran
| | - Sirous Ghobadi
- Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
6
|
Wang J, Wang L, Wang Q, Lu Q. Relaxin 2 Suppresses Tumor Growth in Esophageal Squamous Cell Carcinoma EC9706 Cells Through Modulation of the STAT Signaling Pathway. J Biochem Mol Toxicol 2025; 39:e70288. [PMID: 40343386 DOI: 10.1002/jbt.70288] [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: 03/26/2025] [Revised: 04/03/2025] [Accepted: 04/25/2025] [Indexed: 05/11/2025]
Abstract
Esophageal carcinoma is a highly aggressive cancer with limited therapeutic options. Relaxin 2 (RLN2) has been identified as a regulator of various physiological processes, but its role in cancer, particularly in esophageal carcinoma, is less understood. This study investigates the anticancer effects of RLN2 in EC9706 esophageal carcinoma cells and explores its involvement in the STAT signaling pathway. The study utilized various assays, including CCK-8 for cell viability, DAPI staining and annexin V-FITC for apoptosis assessment, transwell chambers for cell migration and invasion, tube formation assay for angiogenesis, and western blot analysis for analyzing apoptosis-related and STAT signaling proteins. Additionally, the in vivo anticancer potential of RLN2 was assessed using a mouse model. RLN2 demonstrated significant cytotoxicity against esophageal EC9706 cancer cells, inhibiting cell proliferation in a dose- and time-dependent manner. Apoptosis was markedly increased with higher doses of RLN2, as evidenced by an increase in both early and late apoptotic cells. Proapoptotic proteins (Bad, Bax, Caspase-3, Caspase-9) were upregulated, while antiapoptotic proteins (Bcl-2, Mcl-1, Bcl-xL) were downregulated in response to RLN2 treatment. RLN2 significantly inhibited cell migration, invasion, and angiogenesis in EC9706 cells. Furthermore, RLN2 increased the phosphorylation of STAT-1, while the phosphorylation of STAT-3 was reduced. In the in vivo model, RLN2 effectively suppressed tumor growth. RLN2 exhibits potent anticancer effects against EC9706 cells and in a mouse model, primarily through inducing intrinsic apoptosis, inhibiting cell migration and invasion, and blocking the STAT signaling pathway. These findings highlight RLN2's potential as a therapeutic agent for esophageal carcinoma.
Collapse
Affiliation(s)
- Juzheng Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Thoracic Surgery, The First People's Hospital of Xianyang, Xianyang, China
| | - Lv Wang
- Department of Thoracic Surgery, Honghui Hospital of Xi'an, Xi'an, China
| | - Qingshi Wang
- Department of Thoracic Surgery, The First People's Hospital of Xianyang, Xianyang, China
| | - Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
7
|
Zhang J, Feng S, Geng Y, Wang X, Wang Z, Liu Y. Anti-inflammatory phenotypes of immune cells after myocardial infarction and prospects of therapeutic strategy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04167-y. [PMID: 40278891 DOI: 10.1007/s00210-025-04167-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025]
Abstract
Often causing negative cardiac remodeling and heart failure, a major threat to human life and health, myocardial infarction (MI) is a cardiovascular disease with a high morbidity and fatality rate worldwide. Maintaining ordinary heart function depends significantly on the immune system. Necrotic cardiomyocyte signals promote specific immunity and activate general immunity as the disease progresses in MI. Complex immune cells play a key role in all stages of MI progression by removing necrotic cardiomyocytes and tissue and promoting the healing of damaged tissue cells. Immune cells can help to regrow injured heart muscle as well as enable both inflammation and cardiomyocyte death. Immune cells are essential elements that help the immune system carry out its protective function. There are two types of immunity: nonspecific immunity and specific immunity. Developed throughout the long-term evolution of species, nonspecific immunity (including macrophages, myeloid-derived suppressor cells MDSC, natural killer cells NK, neutrophils, and dendritic cells DC) offers immediate and conservative host defense that might destroy healthy tissues because of its nonspecific nature. Precisely acquired immunity, specific immunity helps humoral and cellular immunity mediated through B and T cells correspondingly. These findings offer crucial information needed for the creation of effective immunomodulatory treatment, as discussed in this article.
Collapse
Affiliation(s)
- Jiacheng Zhang
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, Shandong, China
| | - Shuai Feng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250300, Shandong, China
| | - Yannan Geng
- Pharmaceutical Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, Shandong, China
| | - Xiaoli Wang
- Pharmaceutical Department, Liaocheng People's Hospital, Liaocheng, 252002, Shandong, China
| | - Zhen Wang
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, Shandong, China.
| | - Yang Liu
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, Shandong, China.
| |
Collapse
|
8
|
Agrawal N, Afzal M, Almalki WH, Ballal S, Sharma GC, Krithiga T, Panigrahi R, Saini S, Ali H, Goyal K, Rana M, Abida Khan. Longevity mechanisms in cardiac aging: exploring calcium dysregulation and senescence. Biogerontology 2025; 26:94. [PMID: 40259024 DOI: 10.1007/s10522-025-10229-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 03/20/2025] [Indexed: 04/23/2025]
Abstract
Cardiac aging is a multistep process that results in a loss of various structural and functional heart abilities, increasing the risk of heart disease. Since its remarkable discovery in the early 1800s, when limestone is heated, calcium's importance has been defined in numerous ways. It can help stiffen shells and bones, function as a reducing agent in chemical reactions, and play a central role in cellular signalling. The movement of calcium ions in and out of cells and between those is referred to as calcium signalling. It influences the binding of the ligand, enzyme activity, electrochemical gradients, and other cellular processes. Calcium signalling is critical for both contraction and relaxation under the sliding filament model of heart muscle. However, with age, the heart undergoes changes that lead to increases in cardiac dysfunction, such as myocardial fibrosis, decreased cardiomyocyte function, and noxious disturbances in calcium homeostasis. Additionally, when cardiac tissues age, cellular senescence, a state of irreversible cell cycle arrest, accumulates and begins to exacerbate tissue inflammation and fibrosis. This review explores the most recent discoveries regarding the role of senescent cell accumulation and calcium signalling perturbances in cardiac aging. Additionally, new treatment strategies are used to reduce aged-related heart dysfunction by targeting senescent cells and modulating calcium homeostasis.
Collapse
Affiliation(s)
- Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - T Krithiga
- Department of Chemistry, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Rajashree Panigrahi
- Department of Microbiology IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Suman Saini
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248007, India
| | - Abida Khan
- Center For Health Research, Northern Border University, Arar 73213, Saudi Arabia
| |
Collapse
|
9
|
Tan C, Yuan Z, Xu F, Xie D. Optimized Feature Selection and Deep Neural Networks to Improve Heart Disease Prediction. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025:10.1007/s10278-025-01435-4. [PMID: 40240654 DOI: 10.1007/s10278-025-01435-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 04/18/2025]
Abstract
Heart disease remains a significant health threat due to its high mortality rate and increasing prevalence. Early prediction using basic physical markers from routine exams is crucial for timely diagnosis and intervention. However, manual analysis of large datasets can be labor-intensive and error-prone. Our goal is to rapidly and reliably anticipate cardiac disease using a variety of body signs. This research presents a unique model for heart disease prediction. We provide a system for predicting cardiac disease that blends the deep convolutional neural network with a feature selection technique based on the LinearSVC. This integrated feature selection method selects a subset of characteristics that are strongly linked with heart disease. We feed these features into the deep conventual neural network that we constructed. Also to improve the speed of the predictor and avoid gradient varnishing or explosion, the network's hyperparameters were tuned using the random search algorithm. The proposed method was evaluated using the UCI and MIT datasets. The predictor is evaluated using a number of indicators, such as accuracy, recall, precision, and F1 score. The results demonstrate that our model attains accuracy rates of 98.16%, 98.2%, 95.38%, and 97.84% in the UCI dataset, with an average MCC score of 90%. These results affirm the efficacy and reliability of the proposed technique to predict heart disease.
Collapse
Affiliation(s)
- Changming Tan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, No139 Renmin Road, Changsha, Hunan Province, 410011, People's Republic of China.
| | - Zhaoshun Yuan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, No139 Renmin Road, Changsha, Hunan Province, 410011, People's Republic of China
| | - Feng Xu
- Department of Endocrinology and Metabolism, The Second Xiangya Hospital of Central South University, No139 Renmin Road, Changsha, Hunan Province, 410011, People's Republic of China
| | - Dang Xie
- Weimu (Shanghai) Medical Technology Ltd. No, 4188, Canghai Road, Lingang New Area, Shanghai Free Trade Zone, Shanghai, 201306, China
| |
Collapse
|
10
|
Guo J, Liao J, Chen Y, Wen L, Cheng S. New Machine Learning Method for Medical Image and Microarray Data Analysis for Heart Disease Classification. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025:10.1007/s10278-025-01492-9. [PMID: 40169470 DOI: 10.1007/s10278-025-01492-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/09/2025] [Accepted: 03/19/2025] [Indexed: 04/03/2025]
Abstract
Microarray technology has become a vital tool in cardiovascular research, enabling the simultaneous analysis of thousands of gene expressions. This capability provides a robust foundation for heart disease classification and biomarker discovery. However, the high dimensionality, noise, and sparsity of microarray data present significant challenges for effective analysis. Gene selection, which aims to identify the most relevant subset of genes, is a crucial preprocessing step for improving classification accuracy, reducing computational complexity, and enhancing biological interpretability. Traditional gene selection methods often fall short in capturing complex, nonlinear interactions among genes, limiting their effectiveness in heart disease classification tasks. In this study, we propose a novel framework that leverages deep neural networks (DNNs) for optimizing gene selection and heart disease classification using microarray data. DNNs, known for their ability to model complex, nonlinear patterns, are integrated with feature selection techniques to address the challenges of high-dimensional data. The proposed method, DeepGeneNet (DGN), combines gene selection and DNN-based classification into a unified framework, ensuring robust performance and meaningful insights into the underlying biological mechanisms. Additionally, the framework incorporates hyperparameter optimization and innovative U-Net segmentation techniques to further enhance computational performance and classification accuracy. These optimizations enable DGN to deliver robust and scalable results, outperforming traditional methods in both predictive accuracy and interpretability. Experimental results demonstrate that the proposed approach significantly improves heart disease classification accuracy compared to other methods. By focusing on the interplay between gene selection and deep learning, this work advances the field of cardiovascular genomics, providing a scalable and interpretable framework for future applications.
Collapse
Affiliation(s)
- Jinglan Guo
- Department of Medical Laboratory, Affiliated Hospital of Southwest Medical University, Lu Zhou, 646000, Si Chuan, China
| | - Jue Liao
- School of Basic Medical Sciences of Southwest Medical University, Lu Zhou, 646000, Si Chuan, China
| | - Yuanlian Chen
- Family Planning Service Center, Jiangyang District Maternal and Child Health Hospital, Lu Zhou, 646000, Sichuan, China
| | - Lisha Wen
- Family Planning Service Center, Jiangyang District Maternal and Child Health Hospital, Lu Zhou, 646000, Sichuan, China
| | - Song Cheng
- Department of Medical Laboratory, Affiliated Hospital of Southwest Medical University, Lu Zhou, 646000, Si Chuan, China.
| |
Collapse
|
11
|
Ahmad A, Tiwari RK, Ansari IA. Targeting the interplay between human papillomavirus oncoproteins and hedgehog signaling: assessment of chemopreventive potential of carvacrol in cervical cancer. 3 Biotech 2025; 15:73. [PMID: 40060291 PMCID: PMC11883063 DOI: 10.1007/s13205-025-04250-4] [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: 10/16/2024] [Accepted: 02/18/2025] [Indexed: 04/13/2025] Open
Abstract
Cervical carcinoma is the fourth most frequently diagnosed cancer and is a serious cause of increased mortality among females globally. Hedgehog/GLI signaling has now been established to play a pivotal role in imparting tumor recurrence and promoting metastasis in cervical carcinoma. HPV associated oncoproteins particularly E6/E7 concomitantly with altered signaling pathways are key determinants of cervical cancer. Nevertheless, the nexus between HPV oncogenes and Hedgehog/GLI signaling till date remains unclear. In this study, we investigated the anticancer and apoptotic potential of carvacrol against cervical cancer cells in vitro by targeting the plausible nexus between HPV oncoproteins and Hedgehog signaling. The findings from cell proliferation, LDH cytotoxicity, and morphology analysis suggested that carvacrol treatment significantly decreased the number of viable CaSki cells in a concentration and time-related manner. Morphological trademarks of cell death, including fragmentation of CaSki cell nucleus were studied by DAPI/PI and Hoechst33342 staining. The cytotoxicity of carvacrol was mediated through apoptosis, as confirmed by the Annexin V/FITC assay and caspase activation. Cell cycle analysis showed that carvacrol exerted significant impeding effects on the proliferation of CaSki cells via G0/G1 arrest. Intriguingly, carvacrol mediated the downregulation of HPV E6 and E7 oncogenes indicated its plausible role as an anti-HPV agent against HPV16+ CaSki cells. Additionally, carvacrol further restored p53 expression implicating that carvacrol may protect E6 mediated p53 protein degradation in CaSki cells. Thus, carvacrol exhibited strong antiproliferative potential by inducing apoptosis in cervical carcinoma cells via mediating the crosstalk between the downregulation of HPV oncogenes and inhibition of the hedgehog signaling pathway.
Collapse
Affiliation(s)
- Afza Ahmad
- Present Address: Department of Public Health, Dr. Giri Lal Gupta Institute Public Health and Public Affairs, University of Lucknow, Lucknow, Uttar Pradesh 226031 India
| | - Rohit Kumar Tiwari
- Present Address: Department of Clinical Research, Sharda School of Allied Health Sciences, Sharda University, Gautam Buddha Nagar, Uttar Pradesh 201310 India
| | - Irfan Ahmad Ansari
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh 226026 India
| |
Collapse
|
12
|
Mehta J, Kumar P, Pawar SV. Exploration of capsaicin-encapsulated lignin nanoparticles for alleviating non-alcoholic fatty liver disease: In-vitro study. Int J Biol Macromol 2025; 303:140616. [PMID: 39909244 DOI: 10.1016/j.ijbiomac.2025.140616] [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: 10/10/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a rising public health concern with limited effective therapeutic options, making it a significant risk factor for end-stage liver disease and cirrhosis, globally. This increasing prevalence of NAFLD underscores the development of innovative therapeutic approaches to confront the increasing prevalence of NAFLD. This research explores the potential of capsaicin-encapsulated lignin nanoparticles (Cap-LNPs) as a novel targeted therapeutic approach. Capsaicin, a bioactive compound has demonstrated anti-oxidant, anti-inflammatory, anti-steatotic, and anti-fibrotic properties that have a protective role against NAFLD. Lignin, recognized for its non-toxic, eco-friendly, multifunctional, and biodegradable attributes, has garnered significant attention as a versatile material for drug delivery systems. Incorporating these two natural compounds into nanoparticles offers a promising approach to enhance capsaicin's bioavailability, stability, and targeted delivery to hepatic cells. The Cap-LNPs were synthesized using the nanoprecipitation technique and characterized by a mean diameter of 200.2 ± 5.79 nm, polydispersity index (PDI) of 0.137 ± 0.0459 with spherical morphology, encapsulation efficiency of 96.85 ± 0.73 %, and drug loading capacity of 16.14 ± 0.12 %. In-vitro studies demonstrated that Cap-LNPs substantially reduced intracellular accumulation of triglyceride compared with free capsaicin and control groups, confirmed by Oil Red O staining and triglycerides (TG) quantification.
Collapse
Affiliation(s)
- Jhalak Mehta
- Pharmaceutical Biotechnology Research Lab, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| | - Pankaj Kumar
- Pharmaceutical Biotechnology Research Lab, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| | - Sandip V Pawar
- Pharmaceutical Biotechnology Research Lab, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
13
|
Salehi Namini M, Khanmohammadi M, Beheshtizadeh N, Najafi MS, Heirani-Tabasi A, Ayati A, Boroumand S, Pournemati B, Ai J, Ebrahimi-Barough S, Montazerghaem H, Ahmadi Tafti SH. Injectable hyaluronic acid-based microcapsules loaded with human endometrial stem cells improves cardiac function after myocardial infarction. Int J Biol Macromol 2025; 304:140904. [PMID: 39938851 DOI: 10.1016/j.ijbiomac.2025.140904] [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: 05/01/2024] [Revised: 01/18/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025]
Abstract
Therapeutic efficacy of human endometrial stem cells (hEnSCs) encapsulated in hyaluronic acid (HA)-based microcapsules for cardiac regeneration in a rat model of MI is investigated. Cell-enclosed microcapsules were made by loading hEnSCs within hydrogel membrane produced from modified HA possessing phenolic hydroxyl moieties (HA-Ph). The hEnSC-loaded HA-Ph microcapsules (≈150 μm) injected intramyocardially into the peri-infarct area post-MI. The encapsulated cells showed mechanical stability and >87 % cell viability with cellular aggregation in size of about 100 μm until 7 days of culture. Transthoracic echocardiography evaluation indicated a significant increase in ejection fraction in encapsulated cells, compared to the other groups. Histological investigation of fibrosis and scar area by Masson trichrome and hematoxylin and eosin (H&E) staining illustrated less fibrosis and scarring area in the encapsulated cell group compared with the other groups. Furthermore, the cell-laden microcapsules significantly enhance expression intensities of actin and troponin as well as vascular endothelial-specific marker, all of which promote cardiac functions and contribute to a better therapeutic effect than the free-cell injection group in a rat model of MI. Our findings demonstrated that both hEnSCs and specifically hEnSC-loaded HA-based hydrogel vehicle can provide a promising novel therapy for functional restoration in MI instances.
Collapse
Affiliation(s)
- Mojdeh Salehi Namini
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mehdi Khanmohammadi
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Wołoska 141, Warsaw 02-507, Poland
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Sadeq Najafi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Asieh Heirani-Tabasi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aryan Ayati
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Safieh Boroumand
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnam Pournemati
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Montazerghaem
- Cardiovascular Research center, Hormozgan University of Medical Science, Bandar Abbas, Iran
| | - Seyed Hossein Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Nasiri R, Arefnezhad R, Baniasad K, Hosseini SA, Jeshari AS, Miri M, Lotfi A, Ghaemi MS, Amini-Salehi E, Fatemian H, Rezaei-Tazangi F, Kesharwani P, Tavakoli MR, Sahebkar A. Baicalin and baicalein against myocardial ischemia-reperfusion injury: A review of the current documents. Tissue Cell 2025; 93:102772. [PMID: 39923649 DOI: 10.1016/j.tice.2025.102772] [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: 12/14/2024] [Revised: 01/13/2025] [Accepted: 02/01/2025] [Indexed: 02/11/2025]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a significant challenge in the treatment of ischemic heart disease (IHD), arising as a complication from reperfusion therapies designed to restore blood flow after an ischemic event. Despite the availability of various therapeutic strategies, finding an effective treatment for MIRI remains difficult. Baicalin and its aglycone form (baicalein), natural compounds derived from the Chinese skullcap plant (Scutellaria baicalensis), have shown promise due to their antioxidant, anti-inflammatory, and cardioprotective properties. This review aims to explore the potential of baicalin and baicalein as treatments for MIRI, with a focus on their molecular and cellular level effects. These natural agents can decrease oxidative stress by promoting antioxidant enzymes and decreasing harmful oxidative substances that damage cardiac cells. They also exert anti-inflammatory effects by blocking specific pathways that trigger the release of inflammatory mediators. Additionally, they also improve heart cell survival, infarct region, and overall cardiac function by inhibiting key signaling pathways involved in cell death. Research in both animal and cell models suggests that these flavonoids, especially baicalin, can restore cardiac health following MIRI, improving cardiac performance, and reducing cardiac damage. These findings underscore the potential of baicalin and baicalein as therapeutic options for MIRI. However, further research and clinical trials are necessary to elucidate their mechanisms fully and to develop baicalin into a viable treatment.
Collapse
Affiliation(s)
- Reza Nasiri
- School of Medicine, Shiraz University of Medial Sciences, Shiraz, Iran
| | - Reza Arefnezhad
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Coenzyme R Research Institute, Tehran, Iran
| | - Kimia Baniasad
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
| | - Seyed Ali Hosseini
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mostafa Miri
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Lotfi
- Department of Medical Sciences, School of Medicine, Azerbaijan Medical University, Baku, Azerbaijan
| | - Mozhan Sadat Ghaemi
- Students Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Amini-Salehi
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Hossein Fatemian
- School of Medicine, Shiraz University of Medial Sciences, Shiraz, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Marziye Ranjbar Tavakoli
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Khan A, Alzahrani HA, Felemban SG, Algarni AS, Alenezi ABS, Kamal M, Rehman ZU, Asdaq SMB, Ahmed N, Alharbi BM, Alanazi BS, Imran M. Exploring TGF-β signaling in benign prostatic hyperplasia: from cellular senescence to fibrosis and therapeutic implications. Biogerontology 2025; 26:79. [PMID: 40159577 DOI: 10.1007/s10522-025-10226-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
As men get older, they often develop benign prostatic hyperplasia (BPH), an enlarged prostate that is not cancerous or dangerous. Although the etiology of BPH is unknown, increasing evidence indicates that the TGF-β signaling pathway might be a key player in its pathogenesis. TGF-β is a pleiotropic cytokine involved in proliferation, differentiation, and extracellular matrix re-modeling, which are all dysregulated in BPH. Cellular senescence is primarily initiated by TGF-β--induced, irreversible growth arrest and usually limits the prostate gland's hyperplastic growth. Moreover, senescent cells generate a Senescence-Associated Secretory Phenotype (SASP), which consists of numerous proinflammatory and profibrotic factors that can worsen disease ontogeny. In addition, TGF-β is among the most fibrogenic factors. At the same time, fibrosis involves a massive accumulation of extracellular matrix proteins, which can increase tissue stiffness and a loss of normal organ functions. TGF-β-mediated fibrosis in BPH changes the mechanical properties of the prostate and surrounding tissues to contribute to lower urinary tract symptoms. This review discusses the complicated molecular signaling of TGF-β underlying changes in cellular senescence and fibrosis during BPH concerning its therapeutic potential.
Collapse
Affiliation(s)
- Abida Khan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, 73213, Saudi Arabia
| | - Hayat Ali Alzahrani
- Medical Laboratory Technology Department, College of Medical Applied Science, Northern Border University, Arar, Saudi Arabia
| | - Shatha Ghazi Felemban
- Medical Laboratory Sciences Department, Fakeeh College for Medical Sciences, 21461, Jeddah, Saudi Arabia
| | - Alanood Saeed Algarni
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Zia Ur Rehman
- Health Research Centre, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jazan University, P.O. Box 114, Jazan, 45142, Kingdom of Saudi Arabia
| | | | - Naveed Ahmed
- Department of Assistance Medical Sciences, Applied College, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Bashayer Mohammed Alharbi
- Department of Pharmacy, Johns Hopkins Aramco Healthcare, P.O. Box 10352, 31311, Dhahran, Eastern Province, Saudi Arabia
| | - Bander Sharqi Alanazi
- Department of Nursing Administration, Northern Area Armed Forces Hospital, 31991, Hafer AlBaten, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, 73213, Saudi Arabia.
| |
Collapse
|
16
|
Ribeiro V, dos Santos NSS, de Oliveira RVS, Carvalho J, Garcia VV, Brito-Junior HJS, Usfinit W, Pinheiro M, Fill T, Gester R, Provasi PF, Canuto S, Bitencourt HR, Marinho PSB, Marinho AMR. Synthesis, Biotransformation, Characterization, and DFT Study of Organic Azachalcone Dyes and Secondary Metabolites with Biological and Conformation Dependence of Dipolar-Octupolar NLO Responses. ACS OMEGA 2025; 10:10962-10971. [PMID: 40160730 PMCID: PMC11947782 DOI: 10.1021/acsomega.4c09074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025]
Abstract
Chalcones are organic chromophores with diverse biological applications and potential for use in various electronic devices due to their recognized optical properties. This research focuses on the organic synthesis, FT-NMR characterization, and biotransformation of three azachalcones (1-3) using the Exserohilum rostratum fungus, yielding novel compounds (1a-3a). In vitro biological assays against Gram-positive and Gram-negative bacteria revealed promising pharmacological potential for these new chromophores. A key structural difference, the interchange of an HC = CH bond by a H2C-CH2 bond, significantly impacts biological and electronic properties. For instance, while biotransformed 1a exhibits similar activity to tetracycline and amoxicillin, compounds 2a and 3a demonstrate a 4-fold and thirty-fold increase in inhibitory activity against Gram-negative E. coli, respectively, compared to their parent compounds. Density functional theory calculations suggest that the biotransformation reaction reduces the refractive index (n), which may limit its applicability in certain light-handling applications. However, Hyper-Rayleigh scattering calculations indicate that these chromophores exhibit higher nonlinear optical (NLO) responses compared to standard NLO materials such as urea and p-nitroaniline, making them promising candidates for photonic and optoelectronic devices, such as nanostructured circuits. Interestingly, while the original molecules exhibit a dominant dipolar (Φ J=1) NLO response, the biotransformed compounds, as stable isomers, display a predominant octupolar (Φ J=3) architecture. These findings highlight the potential of these novel compounds for biotechnological and optoelectronic applications.
Collapse
Affiliation(s)
- Victoria
L. Ribeiro
- Programa
de Pós-Graduação em Química, Universidade Federal do Pará, Rua Augusto Corrêa, 01 -
Guamá, 66075-110 Belém, PA, Brazil
| | - Neidy S. S. dos Santos
- Programa
de Pós-Graduação em Química, Universidade Federal do Sul e Sudeste do Pará, 68507-590 Marabá, PA, Brazil
| | - Raira V. S. de Oliveira
- Programa
de Pós-Graduação em Química, Universidade Federal do Sul e Sudeste do Pará, 68507-590 Marabá, PA, Brazil
| | - Joselina
A. Carvalho
- Programa
de Pós-Graduação em Química, Universidade Federal do Sul e Sudeste do Pará, 68507-590 Marabá, PA, Brazil
| | - Viviane V. Garcia
- Programa
de Pós-Graduação em Química, Universidade Federal do Sul e Sudeste do Pará, 68507-590 Marabá, PA, Brazil
| | - Hartmann J. S. Brito-Junior
- Programa
de Pós-Graduação em Química, Universidade Federal do Pará, Rua Augusto Corrêa, 01 -
Guamá, 66075-110 Belém, PA, Brazil
| | - Willibrodus Usfinit
- Programa
de Pós-Graduação em Química, Universidade Federal do Pará, Rua Augusto Corrêa, 01 -
Guamá, 66075-110 Belém, PA, Brazil
| | - Mayra Pinheiro
- Instituto
de Química, Universidade de Campinas, 13083-970 Campinas, SP, Brazil
| | - Taicia Fill
- Instituto
de Química, Universidade de Campinas, 13083-970 Campinas, SP, Brazil
| | - Rodrigo Gester
- Faculdade
de Física, Universidade Federal do
Sul e Sudeste do Pará, 68507-590 Marabá, PA, Brazil
- Instituto
de Física, Universidade de São
Paulo, Rua do Matão
1371, 05508-090 São Paulo, SP, Brazil
| | - Patricio F. Provasi
- Department
of Physics, IMIT, Northeastern University, CONICET, AV. Libertad 5500, W 3404 AAS Corrientes, Argentina
| | - Sylvio Canuto
- Instituto
de Física, Universidade de São
Paulo, Rua do Matão
1371, 05508-090 São Paulo, SP, Brazil
| | - Heriberto R. Bitencourt
- Programa
de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Pará, Rua Augusto Corrêa, 01 - Guamá, 66075-110 Belém, PA, Brazil
| | - Patricia S. B. Marinho
- Programa
de Pós-Graduação em Química, Universidade Federal do Pará, Rua Augusto Corrêa, 01 -
Guamá, 66075-110 Belém, PA, Brazil
| | - Andrey M. R. Marinho
- Programa
de Pós-Graduação em Química, Universidade Federal do Pará, Rua Augusto Corrêa, 01 -
Guamá, 66075-110 Belém, PA, Brazil
| |
Collapse
|
17
|
Shah A, Singh D, Mohamed HG, Bharany S, Rehman AU, Hussen S. Electrocardiogram analysis for cardiac arrhythmia classification and prediction through self attention based auto encoder. Sci Rep 2025; 15:9230. [PMID: 40097668 PMCID: PMC11914083 DOI: 10.1038/s41598-025-93906-5] [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: 09/23/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Sudden cardiac arrest among young people is a recent worldwide risk, and it is noticed that people with cardiac arrhythmia are more susceptible to various heart diseases. Manual classification can be error-prone, and certainly, there is a need for automation to classify ECG signals to predict cardiac arrhythmia accurately. The proposed self-attention artificial intelligence auto-encoder algorithm proved an effective cardiac arrhythmia classification strategy with a novel modified Kalman filter pre-processing. We achieved 24.00 SNRimp, 0.055 RMSE, 22.1 PRD% for -5db, 20.4 SNRimp, 0.0245 RMSE, 12 PRD% whereas 14.05 SNRimp, 0.010 RMSE, and 7.25 PRD%, which reduces the ECG signal noise during the pre-processing and improves the visibility of the QRS complex and R-R peaks of ECG waveform. The extracted features were used in network of neurons to execute the classification for MIT-BIH arrhythmia databases using the newly developed self-attention autoencoder (AE) algorithm. The results are compared with existing models, revealing that the proposed system outperforms the classification and prediction of cardiac arrhythmia with a precision of 99.91%, recall of 99.86%, and accuracy of 99.71%. It is confirmed that self-attention-AE training results are promising, and it benefits the diagnosis of ECGs for complex cardiac conditions to solve real-world heart problems.
Collapse
Affiliation(s)
- Ameet Shah
- School of Computer Science and Engineering, Lovely Professional University, Grand Trunk Rd, Phagwara, 144411, Punjab, India
| | - Dhanpratap Singh
- School of Computer Science and Engineering, Lovely Professional University, Grand Trunk Rd, Phagwara, 144411, Punjab, India.
| | - Heba G Mohamed
- Department of Electrical Engineering, College of Engineering , Princess Nourah bint Abdulrahman University, Riyadh, P.O. Box 84428, 11671, Saudi Arabia.
| | - Salil Bharany
- Chitkara University Institute of Engineering and Technology , Chitkara University, Rajpura, 140401, Punjab, India
| | - Ateeq Ur Rehman
- Computer Science and Engineering, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamilnadu, India
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
- University Center for Research and Development, Chandigarh University, Mohali, 140413, Punjab, India
| | - Seada Hussen
- Department of Electrical Power, Adama Science and Technology University, Adama, 1888, Ethiopia.
| |
Collapse
|
18
|
Rana JN, Mumtaz S. Prunin: An Emerging Anticancer Flavonoid. Int J Mol Sci 2025; 26:2678. [PMID: 40141319 PMCID: PMC11942023 DOI: 10.3390/ijms26062678] [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: 02/13/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Despite the substantial advances in cancer therapies, developing safe and effective treatment methodologies is critical. Natural (plant-derived compounds), such as flavonoids, might be crucial in developing a safe treatment methodology without toxicity toward healthy tissues. Prunin is a flavonoid with the potential to be used in biomedical applications. Prunin has yet to undergo thorough scientific research, and its precise molecular mechanisms of action remain largely unexplored. This review summarizes the therapeutic potential of prunin for the first time, focusing on its underlying mechanisms as an anticancer compound. Prunin has gained significant attention due to its antioxidant, anti-inflammatory, and anticancer effects. This review aims to unlock how prunin functions at the molecular level to exert its anticancer effects, primarily modulating key cellular pathways. Furthermore, we have discussed the prunin's potential as an adjunctive therapy with conventional treatments, highlighting its ability to strengthen treatment responses while decreasing drug resistance. Moreover, the discussion probes into innovative delivery methods, particularly nanoformulations, that might address prunin's bioavailability, solubility, and stability limitations and optimize its therapeutic application. By providing a comprehensive analysis of prunin's properties, this review aims to stimulate further exploration of using prunin as an anticancer agent, thereby progressing the development of targeted, selective, safe, and effective therapeutic methods.
Collapse
Affiliation(s)
- Juie Nahushkumar Rana
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Sohail Mumtaz
- Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnamdaero, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| |
Collapse
|
19
|
Wang C, Du Y, Lu C, Bi L, Ding Y, Fan W. The role of SFRP1 in human dermal papilla cell growth and its potential molecular mechanisms as a target in regenerative therapy. Regen Ther 2025; 28:161-168. [PMID: 39802633 PMCID: PMC11718413 DOI: 10.1016/j.reth.2024.12.001] [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: 11/01/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
Background Secreted frizzled-related protein 1 (SFRP1) inhibits Wnt signaling and is differentially expressed in human hair dermal papilla cells (DPCs). However, the specific effect of SFRP1 on cell function remains unclear. Telomerase reverse transcriptase (TERT) representing telomerase activity was found highly active around the hair dermal papilla. TERT levels can be enhanced by activation of the Wnt pathway in cancer cells and embryonic stem cells. Whether this regulatory mechanism is still present in DPCs has not been studied so far. Methods In this study, DNA plasmids and siRNAs were constructed against the SFRP1 gene and transfected into DPCs cultured in vitro. We detected the viability, proliferation, and migration of DPCs by Calcein/PI fluorescence, CCK-8, trans-well, or cell scratch experiments, and the expression of potential target genes was also determined through quantitative detection of RNA and protein. Results The results demonstrate a significant difference in SFRP1 levels from the control group, suggesting successful transfection of the DNA plasmid and siRNA of SFRP1 into IDPCs. Also, SFRP1 regulates the cell proliferation capacity of IDPCs and reduces their migration functions. The DPCs' living activity, proliferation, and migration function exhibited a negative correlation with the level of SFRP1. SFPR1 also inhibits the protein or RNA expression of β-catenin and TERT in DPCs. Conclusion It was proven that in human DPCs, different levels of SFRP1 change how cells work and control Wnt/β-catenin signaling or telomerase activity. This means that blocking SFRP1 could become a new way to treat hair loss diseases in the future.
Collapse
Affiliation(s)
- Chaofan Wang
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210000, Jiangsu, China
| | - Yimei Du
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Changpei Lu
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Lingbo Bi
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Yunbu Ding
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Weixin Fan
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| |
Collapse
|
20
|
Karami E, Bazgir B, Shirvani H, Mohammadi MT, Khaledi M. Unraveling the bidirectional relationship between muscle inflammation and satellite cells activity: influencing factors and insights. J Muscle Res Cell Motil 2025; 46:35-51. [PMID: 39508952 DOI: 10.1007/s10974-024-09683-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Inflammation stands as a vital and innate function of the immune system, essential for maintaining physiological homeostasis. Its role in skeletal muscle regeneration is pivotal, with the activation of satellite cells (SCs) driving the repair and generation of new myofibers. However, the relationship between inflammation and SCs is intricate, influenced by various factors. Muscle injury and repair prompt significant infiltration of immune cells, particularly macrophages, into the muscle tissue. The interplay of cytokines and chemokines from diverse cell types, including immune cells, fibroadipogenic progenitors, and SCs, further shapes the inflammation-SCs dynamic. While some studies suggest heightened inflammation associates with reduced SC activity and increased fibro- or adipogenesis, others indicate an inflammatory stimulus benefits SC function. Yet, the existing literature struggles to delineate clearly between the stimulatory and inhibitory effects of inflammation on SCs and muscle regeneration. This paper comprehensively reviews studies exploring the impact of pharmacological agents, dietary interventions, genetic factors, and exercise regimes on the interplay between inflammation and SC activity.
Collapse
Affiliation(s)
- Esmail Karami
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Bazgir
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hossein Shirvani
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Mohammadi
- Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mansoor Khaledi
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
21
|
Jin Y, Lai Y, Hoshyar AN, Innab N, Shutaywi M, Deebani W, Swathi A. An ideally designed deep trust network model for heart disease prediction based on seagull optimization and Ruzzo Tompa algorithm. Sci Rep 2025; 15:6035. [PMID: 39971944 PMCID: PMC11839990 DOI: 10.1038/s41598-025-89348-8] [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: 03/30/2024] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
Diet, stress, genetics, and a sedentary lifestyle may all contribute to heart disease rates. Although recent studies propose comprehensive automated diagnostic systems, these systems tend to focus on one aspect, such as feature selection, prioritization, or predictive accuracy. A more complete approach that considers all of these factors can improve the efficiency of a cardiac prediction system. This study uses an appropriate strategy to overcome potential network design problems, design challenges, overfitting, and lack of robustness that can interfere with system performance. The research introduces an ideally designed deep trust network called ID-DTN to improve system performance. The Ruzzo-Tompa method is used to eliminate noncontributory features. The Seagull Optimization Algorithm (SOA) is introduced to optimize the trust depth network to achieve optimal network design. The study scrutinizes the deep trust network (ID-DTN) and the restricted Boltzmann machine (RBM) and sheds light on the system's operation. This proposal can optimize both network architecture and feature selection, which is the main novelty. The proposed method is analyzed using the below-mentioned metrics: Matthew's correlation coefficient, F1 score, accuracy, sensitivity, specificity, and accuracy. ID-DTN performs well compared to other state-of-the-art methods. The validation results confirm that the proposed method improves the prediction accuracy to 97.11% and provides reliable recommendations for patients with cardiovascular disease.
Collapse
Affiliation(s)
- Yuan Jin
- Department of Cardiovascular Medicine, The Fifth People's Hospital of Ganzhou, Ganzhou, 341000, Jiangxi, China.
- Department of Internal Medicine, Xingguo County People's Hospital, Ganzhou, 341000, Jiangxi, China.
| | - Yunliang Lai
- Department of Cardiovascular Medicine, The Fifth People's Hospital of Ganzhou, Ganzhou, 341000, Jiangxi, China
- Department of Internal Medicine, Xingguo County People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Azadeh Noori Hoshyar
- Institute of Innovation, Science and Sustainability, Federation University Australia, Berwick, VIC, 3806, Australia
| | - Nisreen Innab
- Department of Computer Science and Information Systems, College of Applied Sciences, AlMaarefa University, 13713, Diriyah, Riyadh, Saudi Arabia
| | - Meshal Shutaywi
- Department of Mathematics, College of Science and Arts, King Abdul Aziz University, Rabigh, Saudi Arabia
| | - Wejdan Deebani
- Department of Mathematics, College of Science and Arts, King Abdul Aziz University, Rabigh, Saudi Arabia
| | - A Swathi
- Department of Mathematics, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic
| |
Collapse
|
22
|
Yu Z, Kawashima N, Sunada-Nara K, Wang S, Han P, Kieu TQ, Ren C, Noda S, Tazawa K, Okiji T. MicroRNA-27a transfected dental pulp stem cells undergo odonto/osteogenic differentiation via targeting DKK3 and SOSTDC1 in Wnt/BMP signaling in vitro and enhance bone formation in vivo. J Transl Med 2025; 23:189. [PMID: 39956898 PMCID: PMC11831854 DOI: 10.1186/s12967-025-06208-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/05/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) play a crucial role in cell differentiation through epigenetic regulation of gene expression. In human dental pulp cells, we have identified miRNA-27a being upregulated under inflammatory conditions. Here, we aimed to examine whether (i) overexpression of miRNA-27a in human dental pulp stem cells (hDPSCs) enhances their odonto/osteoblastic differentiation via Wnt and bone morphogenetic protein signaling; and (ii) hDPSCs overexpressing miRNA-27a promote new bone formation in vivo. METHODS hDPSCs were cultured in osteogenic medium to promote differentiation. To examine the role of miRNA-27a, hDPSCs were transfected with either a miRNA-27a mimic to enhance or an inhibitor to suppress miRNA-27a expression. Odonto/osteoblastic differentiation was assessed by evaluating the expression of specific markers, Wnt and bone morphogenetic protein (BMP) signaling molecules, and mineralization capacity using RT-qPCR, western blotting, Alizarin Red S (ARS) staining, and alkaline phosphatase (ALP) activity. Potential miRNA-27a binding sites in the 3'UTRs of DKK3 and SOSTDC1 were identified via bioinformatics analysis and validated through the luciferase reporter assay. In vivo, miRNA-27a-overexpressing hDPSCs were seeded into collagen honeycomb scaffolds and implanted into mouse calvarial bone cavities to assess new bone formation. RESULTS MiRNA-27a was highly upregulated in hDPSCs committed to odonto/osteoblastic differentiation. Overexpression of miRNA-27a led to increased expression of odonto/osteoblastic markers and enhanced mineralization capacity, while inhibition of miRNA-27a had the opposite effect. MiRNA-27a targeted DKK3, promoting β-catenin nuclear translocation and inhibiting SOSTDC1, which enhanced SMAD1/5 phosphorylation. Binding sites for miRNA-27a were identified in the 3'UTRs of DKK3 and SOSTDC1. In vivo, miRNA-27a-overexpressing hDPSCs promoted new bone formation in mouse calvaria bone cavities. CONCLUSION Transfection of miRNA-27a in hDPSCs enhanced their odonto/osteoblastic differentiation by targeting DKK3 and SOSTDC1, thereby promoting the Wnt and BMP signaling. Transplantation of miRNA-27a-overexpressing hDPSCs promoted new bone formation in vivo. These findings deepen our understanding of the effects of miRNA on Wnt and BMP pathways and suggest a potential clinical application for miRNA-27a in promoting hard tissue regeneration, offering a promising therapeutic target for dental and craniofacial tissue reconstruction.
Collapse
Affiliation(s)
- Ziniu Yu
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Formerly Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Nobuyuki Kawashima
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Formerly Tokyo Medical and Dental University, Tokyo, 113-8549, Japan.
| | - Keisuke Sunada-Nara
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Formerly Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Shihan Wang
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Formerly Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Peifeng Han
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Formerly Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Thoai Quoc Kieu
- Department of Pediatric Dentistry, Faculty of Odonto-Stomatology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 17000, Vietnam
| | - Chunmei Ren
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Formerly Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Sonoko Noda
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Formerly Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Kento Tazawa
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Formerly Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Takashi Okiji
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Formerly Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| |
Collapse
|
23
|
Kitagawa T. Hydrothermal treatment of yeast cell wall generates potent anti-proliferative agents targeting MCF7 breast cancer cells effectively even under culture conditions separated by a plastic wall. PLoS One 2025; 20:e0313379. [PMID: 39951420 PMCID: PMC11828357 DOI: 10.1371/journal.pone.0313379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
Traditionally, the yeast cell wall (YCW) has limited applications because of its low solubility. To overcome this, a novel method was developed using a hydrothermal reaction to enhance its solubility and decrease its viscosity; this resulted in the production of a soluble form of YCW, known as the YCW treated with hydrothermal reaction (YCW-H), with broader chemical composition. However, the biological impact of YCW-H is unclear, excluding its reported plant growth-promotion by effectively regulating soil microspheres. This study investigated the potential of YCW-H to inhibit MCF-7 breast cancer cell proliferation. YCW-H demonstrated significant anti-proliferative effects on MCF7 cells, reducing cell growth by 58.7% ± 6.9 even when physically separated from the cells by a plastic wall. The observation suggests the presence of a diffusible factor against cell proliferation in YCW-H, a phenomenon not observed in the presence of untreated YCW. Reactive carbon species (RCS) generated during the hydrothermal treatment of YCW could be responsible for the effect. The addition of Fe(III) ions into YCW-H further amplified RCS production and elevated its inhibitory activity by about 10% across the plastic barrier. Radical adduct concentration of H2O in a tube which was incubated in YCW-H was 0.47 μmol/L, indicating that radicals migrated into the water through the plastic wall. The concentration of radical adducts in H2O in a tube exposed to YCW-H with Fe(III) ions further increased to 0.51 μmol/L, indicating that the growth inhibition was correlated with the increased RCS levels. Furthermore, flow cytometry analysis revealed the cytotoxic effects of YCW-H, indicating YCW-H is applicable to cancer therapy. Therefore, the findings highlight the pivotal role of RCS in the YCW-H anti-cancer activity, suggesting its potential as a promising candidate for the development of novel medical devices for cancer treatment.
Collapse
Affiliation(s)
- Takanori Kitagawa
- Agri Division and R&D Department, ASAHI BIOCYCLE CO.,LTD, Shibuya-ku, Tokyo, Japan
| |
Collapse
|
24
|
Rapuru R, Begum RF, Singh SA, Vellapandian C, Ali N, AlAsmari AF, Prajapati BG. Exploring the therapeutic potential of leriodenine and nuciferine from Nelumbo nucifera for renal fibrosis: an In-silico analysis. Z NATURFORSCH C 2025:znc-2024-0229. [PMID: 39925105 DOI: 10.1515/znc-2024-0229] [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: 10/16/2024] [Accepted: 01/18/2025] [Indexed: 02/11/2025]
Abstract
A major problem in chronic kidney illnesses is renal fibrosis. This research investigates the therapeutic potential of compounds derived from Nelumbo nucifera (Lotus). Comprehensive screening identified these compounds, which exhibit promising binding affinities with key targets associated with renal fibrosis. Leriodenine and Nuciferine demonstrate substantial potential by modulating critical targets such as PTGS2, JUN, EGFR, STAT3, mTOR, and AKT1. The identified biomolecule-target-pathway network highlights the intricate interactions underlying the therapeutic effects of lotus seed compounds in renal fibrosis. Strong binding affinities with PTGS2-PDBID:5F19, Leriodenine -8.99 kcal/mol and Nuciferine -9.33 kcal/mol, and JUN-PDBID:1S9K, Leriodenine -7.95 kcal/mol and Nuciferine -7.05 kcal/mol are shown by molecular docking investigations, indicating their potential as fibrotic process inhibitors. During 10 ns of molecular docking simulations, these compounds demonstrated robust hydrogen-bonding connections within the protein's active site, leading to a possible alteration in the conformation of the ligand-binding site. The research establishes the foundation for future experimental validation, clinical trials, to bridge the translational gap. The research combines target prediction, protein-protein interaction studies, and biomolecular screening to clarify the molecular pathways behind renal fibrosis. We also carried out Insilico molecular docking and carried out molecular dynamics simulation of the best compound identified to obtain more precise results.
Collapse
Affiliation(s)
- Rushendran Rapuru
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603 203, Chengalpattu, Tamil Nadu, India
| | - Rukaiah Fatma Begum
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, Uttar Pradesh, India
| | - S Ankul Singh
- Department of Pharmacology, Faculty of Pharmacy, Dr. M.G.R Educational and Research Institute, Velappanchavadi, Chennai 600 077, Tamil Nadu, India
| | - Chitra Vellapandian
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603 203, Chengalpattu, Tamil Nadu, India
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdullah F AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Bhupendra G Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, 79233 Ganpat University , Kherva, Mahesana, 384012, Gujarat, India
- Faculty of Pharmacy, Silpakorn University, Sanam Chandra Palace Campus,6 Rajamankha Nai Road, Amphoe Muang, Nakhon Pathom Province 73000, Thailand
| |
Collapse
|
25
|
Balaraman AK, Moglad E, Afzal M, Babu MA, Goyal K, Roopashree R, Kaur I, Kumar S, Kumar MR, Chauhan AS, Hemalatha S, Gupta G, Ali H. Liquid biopsies and exosomal ncRNA: Transforming pancreatic cancer diagnostics and therapeutics. Clin Chim Acta 2025; 567:120105. [PMID: 39706249 DOI: 10.1016/j.cca.2024.120105] [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: 11/16/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Pancreatic cancer is a highly fatal malignancy due to poor early detection rate and resistance to conventional therapies. This review examines the potential for liquid biopsy as a transformative technology to identify diagnostic and therapeutic targets in pancreatic cancer. Specifically, we explore emerging biomarkers such as exosomal non-coding RNAs (ncRNAs), circulating tumor DNA (ctDNA), and circulating tumor cells (CTCs). Tumor-derived exosomes contain nucleic acid and protein that reflect the unique molecular landscape of the malignancy and can serve as an alternative diagnostic approach vs traditional biomarkers like CA19-9. Herein we highlight exosomal miRNAs, lncRNAs, and other ncRNAs alongside ctDNA and CTC-based strategies, evaluating their combined ability to improve early detection, disease monitoring and treatment response. Furthermore, the therapeutic implications of ncRNAs such as lncRNA UCA1 and miR-3960 in chemoresistance and progression are also discussed via suppression of EZH2 and PTEN/AKT pathways. Emerging therapeutic strategies that target the immune response, epithelial-mesenchymal transition (EMT) and drug resistance are explored. This review demonstrates a paradigm shift in pancreatic cancer management toward personalized, less invasive and more effective approaches.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - MRavi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, India
| | - S Hemalatha
- Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Porur, Chennai, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| |
Collapse
|
26
|
Gang R, Okello D, Ban Y, Kang Y. A systematic review of Aspilia africana (Pers.) C.D. adams traditional medicinal uses, phytoconstituents, bioactivities, and toxicities. Pharmacol Res 2025; 212:107590. [PMID: 39778640 DOI: 10.1016/j.phrs.2025.107590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/17/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
Aspilia africana (Pers.) C. D. Adams, popularly referred to as wild sunflower, has been used for generations across several African communities to treat various diseases, including malaria, wounds, osteoporosis, diabetes mellitus, gastric ulcers, measles, tuberculosis, stomach ache, rheumatic pains, and gonorrhea. This study aimed to systematically and critically compile data on the traditional medicinal uses, phytochemistry, bioactivities, botanical descriptions, and toxicities of A. africana. Relevant research findings were retrieved and organized from various databases, including PubMed and ScienceDirect, in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. To date, 149 phytochemicals have been identified from various A. africana parts and they primarily belong to the classes of terpenoids, lipids, hydrocarbons, phenolics, and esters. The extracts and bioactive phytochemicals of A. africana have revealed several pharmacological properties, including antimalarial, anticancer, wound healing, anti-inflammatory, antidiabetic, and antimicrobial activities. However, the major components responsible for these bioactivities and their mechanisms of action in some diseases have not yet been clearly identified. Additionally, toxicity and clinical trial data for A. africana are limited with most toxicological assessments being acute in nature. Therefore, further research on the mechanisms of action of the pure bioactive phytochemicals and toxicity of A. africana are necessary to better understand its efficacy and safety. Taken together, this study provides comprehensive information on the traditional medicinal uses, phytochemistry, bioactivities, and toxicity of A. africana, and a reference for future studies, relevant to the development of therapeutic products.
Collapse
Affiliation(s)
- Roggers Gang
- Korean Convergence Medical Science Major, Korea National University of Science and Technology (UST), Daejeon 34113, South Korea; Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine (KIOM), 111 Geonjae-Ro, Naju-Si, South Korea; National Agricultural Research Organization (NARO), National Semi-Arid Resources Research Institute (NaSARRI), Soroti, Uganda
| | - Denis Okello
- Department of Biological Sciences, Kabale University, PO Box 317, Kabale, Uganda
| | - Yeongjun Ban
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine (KIOM), 111 Geonjae-Ro, Naju-Si, South Korea
| | - Youngmin Kang
- Korean Convergence Medical Science Major, Korea National University of Science and Technology (UST), Daejeon 34113, South Korea; Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine (KIOM), 111 Geonjae-Ro, Naju-Si, South Korea.
| |
Collapse
|
27
|
Balaraman AK, Babu MA, Moglad E, Mandaliya V, Rekha MM, Gupta S, Prasad GVS, Kumari M, Chauhan AS, Ali H, Goyal K. Exosome-mediated delivery of CRISPR-Cas9: A revolutionary approach to cancer gene editing. Pathol Res Pract 2025; 266:155785. [PMID: 39708520 DOI: 10.1016/j.prp.2024.155785] [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: 11/13/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Several molecular strategies based on targeted gene delivery systems have been developed in recent years; however, the CRISPR-Cas9 technology introduced a new era of targeted gene editing, precisely modifying oncogenes, tumor suppressor genes, and other regulatory genes involved in carcinogenesis. However, efficiently and safely delivering CRISPR-Cas9 to cancer cells across the cell membrane and the nucleus is still challenging. Using viral vectors and nanoparticles presents issues of immunogenicity, off-target effects, and low targeting affinity. Naturally, extracellular vesicles called exosomes have garnered the most attention as delivery vehicles in oncology-related CRISPR-Cas9 calls due to their biocompatibility, loading capacity, and inherent targeting features. The following review discusses the current progress in using exosomes to deliver CRISPR-Cas9 components, the approaches to load the CRISPR components into exosomes, and the modification of exosomes to increase stability and tumor-targeted delivery. We discuss the latest strategies in targeting recently accomplished in the exosome field, including modifying the surface of exosomes to enhance their internalization by cancer cells, as well as the measures taken to overcome the impacts of TME on delivery efficiency. Focusing on in vitro and in vivo experimentation, this review shows that exosome-mediated CRISPR-Cas9 can potentially treat cancer types, including pancreatic, lymphoma, and leukemia, for given gene targets. This paper compares exosome-mediated delivery and conventional vectors regarding safety, immune response, and targeting ability. Last but not least, we present the major drawbacks and potential development of the seemingly promising field of exosome engineering in gene editing, with references to CRISPR technologies and applications that may help make the target exosomes therapeutic in oncology.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Viralkumar Mandaliya
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Mukesh Kumari
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India.
| |
Collapse
|
28
|
Zhang D, Zhang B. cGAS/STING signaling pathway in gynecological malignancies: From molecular mechanisms to therapeutic values. Front Immunol 2025; 16:1525736. [PMID: 39949780 PMCID: PMC11821648 DOI: 10.3389/fimmu.2025.1525736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Gynecological cancers, including cervical, ovarian, and endometrial malignancies, remain a significant global health burden, exacerbated by disparities in access to preventive measures such as HPV vaccination and routine screening. The cGAS/STING signaling pathway, a pivotal mechanism in innate immunity, detects cytosolic DNA from pathogens or cellular damage, triggering immune responses via type I interferons and inflammatory cytokines. This pathway's dual role in gynecological cancers, either promoting antitumor immunity or facilitating tumor immune evasion, makes it a compelling target for innovative therapies. The article outlines cGAS/STING's influence on tumor microenvironments, immune surveillance, and inflammation, with emphasis on molecular mechanisms driving cancer progression. It explores interactions between DNA damage response pathways and immune modulation, highlighting the impact of cGAS/STING activation or suppression in ovarian, cervical, and endometrial cancers. The therapeutic potential of STING agonists, PARP inhibitors, and targeted immunotherapies is reviewed, demonstrating how these approaches can boost immune responses, counteract chemotherapy resistance, and improve patient outcomes. The study also discusses strategies for leveraging cGAS/STING signaling to enhance the efficacy of immunotherapies and address tumor-mediated immune suppression, providing insights into future directions for personalized cancer treatments.
Collapse
Affiliation(s)
| | - Bingxue Zhang
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
29
|
Wang J, Gao T, Zhang D, Tang Y, Gu J. Phospholipase C epsilon 1 as a therapeutic target in cardiovascular diseases. J Adv Res 2025:S2090-1232(25)00051-7. [PMID: 39855298 DOI: 10.1016/j.jare.2025.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Phospholipase C epsilon 1 (PLCε1) can hydrolyze phosphatidylinositol-4,5-bisphosphate and phosphatidylinositol-4-phosphate at the plasma membrane and perinuclear membrane in the cardiovascular system, producing lipid-derived second messengers. These messengers are considered prominent triggers for various signal transduction processes. Notably, diverse cardiac phenotypes have been observed in cardiac-specific and global Plce1 knockout mice under conditions of pathological stress. It is well established that the cardiac-specific Plce1 knockout confers cardioprotective benefits. Therefore, the development of tissue/cell-specific targeting approaches is critical for advancing therapeutic interventions. AIM OF REVIEW This review aims to distill the foundational biology and functional significance of PLCε1 in cardiovascular diseases, as well as to explore potential avenues for research and the development of novel therapeutic strategies targeting PLCε1. KEY SCIENTIFIC CONCEPTS OF REVIEW Cardiovascular diseases remain the leading cause of morbidity and mortality worldwide, with incidence rates escalating annually. A comprehensive understanding of the multifaceted role of PLCε1 is essential for enhancing the diagnosis, management, and prognostic assessment of patients suffering from cardiovascular diseases.
Collapse
Affiliation(s)
- Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ting Gao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Dongmei Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
30
|
Imran M, Altamimi ASA, Afzal M, Babu MA, Goyal K, Ballal S, Sharma P, Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ali H. Targeting senescence and GATA4 in age-related cardiovascular disease: a comprehensive approach. Biogerontology 2025; 26:45. [PMID: 39831933 DOI: 10.1007/s10522-025-10189-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
The growing prevalence of age-related cardiovascular diseases (CVDs) poses significant health challenges, necessitating the formulation of novel treatment approaches. GATA4, a vital transcription factor identified for modulating cardiovascular biology and cellular senescence, is recognized for its critical involvement in CVD pathogenesis. This review collected relevant studies from PubMed, Google Scholar, and Science Direct using search terms like 'GATA4,' 'cellular senescence,' 'coronary artery diseases,' 'hypertension,' 'heart failure,' 'arrhythmias,' 'congenital heart diseases,' 'cardiomyopathy,' and 'cardiovascular disease.' Additionally, studies investigating the molecular mechanisms underlying GATA4-mediated regulation of GATA4 and senescence in CVDs were analyzed to provide comprehensive insights into this critical aspect of potential treatment targeting. Dysregulation of GATA4 is involved in a variety of CVDs, as demonstrated by both experimental and clinical research, comprising CAD, hypertension, congenital heart diseases, cardiomyopathy, arrhythmias, and cardiac insufficiency. Furthermore, cellular senescence enhances the advancement of age-related CVDs. These observations suggested that therapies targeting GATA4, senescence pathways, or both as necessary may be an effective intervention in CVD progression and prognosis. Addressing age-related CVDs by targeting GATA4 and senescence is a broad mechanism approach. It implies further investigation of the molecular nature of these processes and elaboration of an effective therapeutic strategy. This review highlights the importance of GATA4 and senescence in CVD pathogenesis, emphasizing their potential as therapeutic targets for age-related CVDs.
Collapse
Affiliation(s)
- Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha, 91911, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, UP, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Fadiyah Jadid Alanazi
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Sakaka 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haider Ali
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| |
Collapse
|
31
|
Balaraman AK, Altamimi ASA, Babu MA, Goyal K, PadmaPriya G, Bansal P, Rajotiya S, Kumar MR, Rajput P, Imran M, Gupta G, Thangavelu L. The interplay of senescence and MMPs in myocardial infarction: implications for cardiac aging and therapeutics. Biogerontology 2025; 26:46. [PMID: 39832057 DOI: 10.1007/s10522-025-10190-6] [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: 11/18/2024] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Aging is associated with a marked increase in cardiovascular diseases, such as myocardial infarction (MI). Cellular senescence is also a crucial factor in the development of age-related MI. Matrix metalloproteinases (MMPs) interaction with cellular senescence is a critical determinant of MI development and outcomes, most notably in the aged heart. After experiencing a heart attack, senescent cells exhibit a Senescence-Associated Secretory Phenotype (SASP) and are involved in tissue regeneration and chronic inflammation. MMPs are necessary for extracellular matrix proteolysis and have a biphasic effect, promoting early heart healing and detrimental change if overexpressed shortly. This review analyses the complex connection between senescence and MMPs in MI and how it influences elderly cardiac performance. Critical findings suggest that increasing cellular senescence in aged hearts elevates MMP activity and aggravates extended ventricular remodeling and dysfunction. Additionally, we explore potential therapeutics that address MMPs and senescence to enhance old MI patient myocardial performance and regeneration.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari Cyber 11, Cyberjaya, Selangor, 63000, Malaysia
| | | | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Uttar Pradesh, Mathura, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - G PadmaPriya
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Pooja Bansal
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Sumit Rajotiya
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Pranchal Rajput
- Division of Research and Innovation, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Punjab, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
32
|
Kumar LK, Suma KG, Udayaraju P, Gundu V, Mantena SV, Jagadesh BN. Clustering-based binary Grey Wolf Optimisation model with 6LDCNNet for prediction of heart disease using patient data. Sci Rep 2025; 15:1270. [PMID: 39779935 PMCID: PMC11711402 DOI: 10.1038/s41598-025-85561-7] [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: 12/08/2023] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
In recent years, the healthcare data system has expanded rapidly, allowing for the identification of important health trends and facilitating targeted preventative care. Heart disease remains a leading cause of death in developed countries, often leading to consequential outcomes such as dementia, which can be mitigated through early detection and treatment of cardiovascular issues. Continued research into preventing strokes and heart attacks is crucial. Utilizing the wealth of healthcare data related to cardiac ailments, a two-stage medical data classification and prediction model is proposed in this study. Initially, Binary Grey Wolf Optimization (BGWO) is used to cluster features, with the grouped information then utilized as input for the prediction model. An innovative 6-layered deep convolutional neural network (6LDCNNet) is designed for the classification of cardiac conditions. Hyper-parameter tuning for 6LDCNNet is achieved through an improved optimization method. The resulting model demonstrates promising performance on both the Cleveland dataset, achieving a convergence of 96% for assessing severity, and the echocardiography imaging dataset, with an impressive 98% convergence. This approach has the potential to aid physicians in diagnosing the severity of cardiac diseases, facilitating early interventions that can significantly reduce mortality associated with cardiovascular conditions.
Collapse
Affiliation(s)
- Lella Kranthi Kumar
- School of Computer Science and Engineering, VIT-AP University, Vijayawada, India.
| | - K G Suma
- School of Computer Science and Engineering, VIT-AP University, Vijayawada, India
| | - Pamula Udayaraju
- Department of Computer Science and Engineering, School of Engineering and Sciences, SRM University, Amaravati, AP, India
| | - Venkateswarlu Gundu
- Department of Computer Science and Engineering, Koneru Lakshmaiah Education Foundation, Vaddeswaram, 522302, India
| | - Srihari Varma Mantena
- Department of Computer Science and Engineering, SRKR Engineering College, Bhimavaram, 534204, India
| | - B N Jagadesh
- School of Computer Science and Engineering, VIT-AP University, Vijayawada, India
| |
Collapse
|
33
|
Elshewey AM, Abed AH, Khafaga DS, Alhussan AA, Eid MM, El-Kenawy ESM. Enhancing heart disease classification based on greylag goose optimization algorithm and long short-term memory. Sci Rep 2025; 15:1277. [PMID: 39779779 PMCID: PMC11711398 DOI: 10.1038/s41598-024-83592-0] [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: 11/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Heart disease is a category of various conditions that affect the heart, which includes multiple diseases that influence its structure and operation. Such conditions may consist of coronary artery disease, which is characterized by the narrowing or clotting of the arteries that supply blood to the heart muscle, with the resulting threat of heart attacks. Heart rhythm disorders (arrhythmias), heart valve problems, congenital heart defects present at birth, and heart muscle disorders (cardiomyopathies) are other types of heart disease. The objective of this work is to introduce the Greylag Goose Optimization (GGO) algorithm, which seeks to improve the accuracy of heart disease classification. GGO algorithm's binary format is specifically intended to choose the most effective set of features that can improve classification accuracy when compared to six other binary optimization algorithms. The bGGO algorithm is the most effective optimization algorithm for selecting the optimal features to enhance classification accuracy. The classification phase utilizes many classifiers, the findings indicated that the Long Short-Term Memory (LSTM) emerged as the most effective classifier, achieving an accuracy rate of 91.79%. The hyperparameter of the LSTM model is tuned using GGO, and the outcome is compared to six alternative optimizers. The GGO with LSTM model obtained the highest performance, with an accuracy rate of 99.58%. The statistical analysis employed the Wilcoxon signed-rank test and ANOVA to assess the feature selection and classification outcomes. Furthermore, a set of visual representations of the results was provided to confirm the robustness and effectiveness of the proposed hybrid approach (GGO + LSTM).
Collapse
Affiliation(s)
- Ahmed M Elshewey
- Department of Computer Science, Faculty of Computers and Information, Suez University, P.O.BOX:43221, Suez, Egypt.
| | - Amira Hassan Abed
- Department of Information Systems, High Institution for Marketing, Commerce & Information Systems, Cairo, Egypt
| | - Doaa Sami Khafaga
- Department of Computer Sciences, College of Computer and Information Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Amel Ali Alhussan
- Department of Computer Sciences, College of Computer and Information Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Marwa M Eid
- Faculty of Artificial Intelligence, Delta University for Science and Technology, Mansoura, 11152, Egypt
- Department of Communications and Electronics, Delta Higher Institute of Engineering and Technology, Mansoura, 35111, Egypt
| | - El-Sayed M El-Kenawy
- Department of Communications and Electronics, Delta Higher Institute of Engineering and Technology, Mansoura, 35111, Egypt
- School of ICT, Faculty of Engineering, Design and Information & Communications Technology (EDICT), Bahrain Polytechnic, PO Box 33349, Isa Town, Bahrain
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
- Jadara University Research Center, Jadara University, Irbid, Jordan
| |
Collapse
|
34
|
Qureshi M, Ishaq K, Daniyal M, Iftikhar H, Rehman MZ, Salar SAA. Forecasting cardiovascular disease mortality using artificial neural networks in Sindh, Pakistan. BMC Public Health 2025; 25:34. [PMID: 39754102 PMCID: PMC11699765 DOI: 10.1186/s12889-024-21187-0] [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: 05/10/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death and disability worldwide, and its incidence and prevalence are increasing in many countries. Modeling of CVD plays a crucial role in understanding the trend of CVD death cases, evaluating the effectiveness of interventions, and predicting future disease trends. This study aims to investigate the modeling and forecasting of CVD mortality, specifically in the Sindh province of Pakistan. The civil hospital in the Nawabshah area of Sindh province, Pakistan, provided the data set used in this study. It is a time series dataset with actual cardiovascular disease (CVD) mortality cases from 1999 to 2021 included. This study analyzes and forecasts the CVD deaths in the Sindh province of Pakistan using classical time series models, including Naïve, Holt-Winters, and Simple Exponential Smoothing (SES), which have been adopted and compared with a machine learning approach called the Artificial Neural Network Auto-Regressive (ANNAR) model. The performance of both the classical time series models and the ANNAR model has been evaluated using key performance indicators such as Root Mean Square Deviation Error, Mean Absolute Error (MAE), and Mean Absolute Percentage Error (MAPE). After comparing the results, it was found that the ANNAR model outperformed all the selected models, demonstrating its effectiveness in predicting CVD mortality and quantifying future disease burden in the Sindh province of Pakistan. The study concludes that the ANNAR model is the best-selected model among the competing models for predicting CVD mortality in the Sindh province. This model provides valuable insights into the impact of interventions aimed at reducing CVD and can assist in formulating health policies and allocating economic resources. By accurately forecasting CVD mortality, policymakers can make informed decisions to address this public health issue effectively.
Collapse
Affiliation(s)
- Moiz Qureshi
- Govt Degree College TangoJam, Hyderabad 70060, Sindh, Pakistan
- Department of Statistics, Quaid-i-Azam University, 45320, Islamabad, Pakistan
| | - Khushboo Ishaq
- Ibn-e-Sina Medical University Mirpurkhas, Sindh, Pakistan
| | - Muhammad Daniyal
- Department of Statistics, Faculty of Computing, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Hasnain Iftikhar
- Department of Statistics, Quaid-i-Azam University, 45320, Islamabad, Pakistan.
- Al-Barkaat Institute of Management Studies, Aligarh 202122, Dr. A. P. J. Abdul Kalam Technical University, Lucknow 226010, India.
| | - Mohd Ziaur Rehman
- Department of Finance, College of Business Administration, King Saud University, P.O. Box 71115, Riyadh, 11587, Saudi Arabia
| | - S A Atif Salar
- Al-Barkaat Institute of Management Studies, Aligarh 202122, Dr. A. P. J. Abdul Kalam Technical University, Lucknow 226010, India
| |
Collapse
|
35
|
Namini MS, Beheshtizadeh N, Ebrahimi-Barough S, Ai J. Human endometrial stem cell-derived small extracellular vesicles enhance neurite outgrowth and peripheral nerve regeneration through activating the PI3K/AKT signaling pathway. J Transl Med 2025; 23:6. [PMID: 39754260 PMCID: PMC11699817 DOI: 10.1186/s12967-024-06048-z] [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: 08/02/2024] [Accepted: 12/25/2024] [Indexed: 01/06/2025] Open
Abstract
Nowadays, extracellular vesicles (EVs) such as exosomes participate in cell-cell communication and gain attention as a new approach for cell-free therapies. Recently, various studies have demonstrated the therapeutic ability of exosomes, while the biological effect of human endometrial stem cell (hEnSC)-derived small EVs such as exosomes is still unclear. Herein, we obtained small EVs from hEnSC and indicated that these small EVs activate the vital cell signaling pathway and progress neurite outgrowth in PC-12 cell lines. For this purpose, hEnSC-derived small EVs were extracted by ultracentrifuge and characterized by DLS, SEM, TEM, and western blot. Also, dil-staining of hEnSC-derived small EVs was done to determine the penetration of hEnSC-derived small EVs into PC12 cells. The MTT assay, scratch assay, and western blot assay were applied to PC12 cells that were exposed to different concentrations of small EVs (0, 50, 100, and 150 µg/ml). Our results demonstrated that small EVs significantly increased neurite outgrowth, proliferation, and migration in PC12 cells in a dose-dependent manner. Moreover, the analysis of western blots showed increased expression of the PI3k/AKT signaling pathway in PC12 cells exposed to hEnSC-derived small EVs in a dose-dependent manner. Also, the results of this study indicated that hEnSC-derived small EVs can enhance cell proliferation and migration and promote neural outgrowth by activating the PI3k/AKT signaling pathway. Accordingly, hEnSC-derived small EVs became an effective strategy for cell-free therapies. Altogether, these positive effects make hEnSC-derived small EVs a new efficient approach in regenerative medicine, especially for the cure of neural injury.
Collapse
Affiliation(s)
- Mojdeh Salehi Namini
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Kakar M, Ullah S, Khan A, Nazir S. Evaluation of efficacy of GCSF in reducing neutropenia among carcinoma patients undergoing anti-cancer chemotherapy. A prospective cohort study. PLoS One 2025; 20:e0315435. [PMID: 39746072 PMCID: PMC11695004 DOI: 10.1371/journal.pone.0315435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025] Open
Abstract
The use of granulocyte colony-stimulating factor (GCSF) to control febrile neutropenia (FN) caused by anti-cancer chemotherapy is well documented but it still needs to evaluated with respect to the specific type of cancer and chemotherapeutic agents. The present study evaluates the efficacy of adjunctive GCSF for treating FN after taking anticancer therapy by measuring clinical, hematological and microbiological outcomes. It is a single center study conducted at Hayatabad Medical Complex (HMC), Peshawar, Pakistan. Adult patients of both genders, suffering from different types of sarcomas and taking anticancer chemotherapy were included in the study. The study was conducted between January 2023 and January 2024. Baseline data including demographic data, medication history and hematological evaluation of all the patients was recorded at the time of enrolment. Primary outcomes of the study were the extent of absolute neutrophil count (ANC) recovery, duration and severity of neutropenia (grade IV), period to fever resolution. After the therapy (with and without adjunctive GCSF) clinical outcomes, hematological evaluation and microbiological data was compared and evaluated. All the data was statistically analyzed by SPSS (IBMS, version 20). A total number of 120 patients were investigated out of which data of 109 patients was included. Out of 109 patients, 64 (58.72%) received adjunctive GCSF therapy, and 45 (41.28%) did not receive adjunctive GCSF. Comparison of the data showed that the patients receiving adjunctive GCSF had a significant improvement ANC recovery time, better recovery of fever and patients were free of infections. This study concluded that adjunctive GCSF therapy benefits the patients undergoing anticancer treatment for different types of carcinoma.
Collapse
Affiliation(s)
- Maria Kakar
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Sami Ullah
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Amjad Khan
- Department of Pharmacy, Kohat University of Science and Technology (KUST), Kohat, Pakistan
| | - Shabnam Nazir
- Department of Pharmacy, Kohat University of Science and Technology (KUST), Kohat, Pakistan
| |
Collapse
|
37
|
Goyal K, Afzal M, Altamimi ASA, Babu MA, Ballal S, Kaur I, Kumar S, Kumar MR, Chauhan AS, Ali H, Shahwan M, Gupta G. Chronic kidney disease and aging: dissecting the p53/p21 pathway as a therapeutic target. Biogerontology 2024; 26:32. [PMID: 39725742 DOI: 10.1007/s10522-024-10173-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
Chronic kidney diseases (CKD) are a group of multi-factorial disorders that markedly impair kidney functions with progressive renal deterioration. Aging contributes to age-specific phenotypes in kidneys, which undergo several structural and functional alterations, such as a decline in regenerative capacity and increased fibrosis, inflammation, and tubular atrophy, all predisposing them to disease and increasing their susceptibility to injury while impeding their recovery. A central feature of these age-related processes is the activation of the p53/p21 pathway signaling. The pathway is a key player in cellular senescence, apoptosis, and cell cycle regulation, which are all key to maintaining the health of the kidney. P53 is a transcription factor and a tumor suppressor protein that responds to cell stress and damage. Persistent activation of cell p53 can lead to the expression of p21, an inhibitor of the cell cycle known as a cyclin-dependent kinase. This causes cells to cease dividing and leads to senescence, where cells can no longer increase. The accumulation of senescent cells in the aging kidney impairs kidney function by altering the microenvironment. As the number of senescent cells increases, the capacity of the kidney to recover from injury decreases, accelerating the progression of end-stage renal disease. This article review extensively explores the relationship between the p53/p21 pathway and cellular senescence within an aging kidney and the emerging therapeutic strategies that target it to overcome the impacts of cellular senescence on CKD.
Collapse
Affiliation(s)
- Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | | | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M Ravi Kumar
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab, 140307, India
| | - Ashish Singh Chauhan
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Haider Ali
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Moyad Shahwan
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
38
|
Hussain SA, Ramasamy M, Shaik MR, Shaik B, Deepak P, Thiyagarajulu N, Matharasi Antonyraj AP, Guru A. Inhibition of Oral Biofilms and Enhancement of Anticancer Activity on Oral Squamous Carcinoma Cells Using Caffeine-Coated Titanium Oxide Nanoparticles. Chem Biodivers 2024:e202402476. [PMID: 39715068 DOI: 10.1002/cbdv.202402476] [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: 10/01/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/25/2024]
Abstract
The fungus Candida albicans is a prominent cariogenic fungal agent that works in association with Streptococcus mutans to accelerate the formation of oral cancer and tooth decay. This study evaluates caffeine-encapsulated titanium oxide nanoparticles (CF-TiO2 NPs) for their potential to prevent biofilm formation on teeth and enhance oral anticancer treatment by influencing apoptotic gene regulation. The synthesized CF-TiO2 NPs were characterized using ultraviolet, scanning electron microscopy, energy-dispersive X-ray spectroscopy, and Fourier-transform infrared spectroscopy analyses, and their antioxidant activity was confirmed through free radical quenching studies. Antimicrobial efficacy was assessed using a zone of inhibition test, revealing strong activity against dental pathogens with a minimal inhibitory concentration of 80 µg/mL. Molecular docking using AutoDock explored interactions between CF and biofilm target sites, supporting their inhibitory potential. In vitro cytotoxicity studies on KB cancer cells showed a dose-dependent increase in cytotoxic effects, with CF-TiO2 NPs promoting apoptotic gene upregulation at concentrations of 20-160 µg/mL. CF-TiO2 NPs demonstrated excellent antioxidant, antibacterial, and anticancer properties, showcasing their promise for oral therapeutic applications. This research highlights a novel approach to managing oral infections and associated complications while improving systemic oral health. Notably, this study is the first to report the biofilm-inhibitory and anticancer potential of CF-TiO2 NPs.
Collapse
Affiliation(s)
- Shaik Althaf Hussain
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohankumar Ramasamy
- Department of Chemistry, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, India
- Interdisciplinary Institute of Indian System of Medicine, SRM Institute of Science and Technology, Kattankulathur, India
| | - Mohammed Rafi Shaik
- Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Baji Shaik
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
| | - Paramasivam Deepak
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| | | | - Anahas Perianaika Matharasi Antonyraj
- Department of Research Analytics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|
39
|
Huang J, Chen Y, Zhou L, Ren J, Tian M, Yang Q, Wang L, Wu Y, Wen J, Yang Q. M2a macrophages regulate fibrosis and affect the outcome after stroke via PU.1/mTOR pathway in fibroblasts. Neurochem Int 2024; 173:105674. [PMID: 38184171 DOI: 10.1016/j.neuint.2024.105674] [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: 10/14/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/08/2024]
Abstract
The moderate formation of the fibrotic scar plays an important role in functional recovery after stroke. M2a macrophages have been identified as an important source of early fibrosis after cerebral ischemia. However, the underlying mechanisms by which macrophages interact with fibroblasts in this context remain largely unknown. Therefore, our study aimed to further investigate the potential mechanisms underlying the effects of macrophages on fibroblasts following ischemic stroke. In vitro and in vivo, recombinant rat interleukin 4 (IL4) was used to induce macrophages to polarize into M2a macrophages. In vitro, primary Sprague-Dawley newborn rat meningeal-derived fibroblasts were treated with PU.1 knockdown, the PU.1 inhibitor DB1976 or the mTOR inhibitor rapamycin, which were then co-cultured with M2a macrophage conditioned medium (MCM). In vivo, Sprague-Dawley adult rats were infected with negative control adenoviruses or PU.1-shRNA adenoviruses. Ten days after infection, an injury model of middle cerebral artery occlusion/reperfusion (MCAO/R) was constructed. Subsequently, IL4 was injected intracerebroventricularly to induce M2a macrophages polarization. In vitro, M2a MCM upregulated PU.1 expression and promoted the differentiation, proliferation, migration and extracellular matrix generation of fibroblasts, which could be reversed by treatment with the PU.1 inhibitor DB1976 or PU.1 knockdown. In vivo, PU.1 expression in fibroblasts was increased within ischemic core following MCAO/R, and this upregulation was further enhanced by exposure to IL4. Treatment with IL4 promoted fibrosis, increased angiogenesis, reduced apoptosis and infarct volume, as well as mitigated neurological deficits after MCAO/R, and these effects could be reversed by PU.1 knockdown. Furthermore, both in vivo and in vitro studies showed that IL4 treatment increased the levels of phosphorylated Akt and mTOR proteins, which were markedly decreased by PU.1 knockdown. Additionally, the use of an mTOR inhibitor rapamycin obviously suppressed the migration and differentiation of fibroblasts, and Col1 synthesis. In conclusion, our findings suggest for the first time that M2a macrophages, at least in part, regulate fibrosis and affect the outcome after cerebral ischemic stroke via the PU.1/mTOR signaling pathway in fibroblasts.
Collapse
Affiliation(s)
- Jiagui Huang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The Second People's Hospital of Yibin, Yibin, China
| | - Yue Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiangxia Ren
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingfen Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinghuan Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Youlin Wu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Wen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
40
|
Xiao Y, Huang Z, Wang Y, Wang Y, Yu L, Yang J, Zou H, Wan W, Yang X. Xanthohumol attenuates collagen synthesis in scleroderma skin fibroblasts by ROS/Nrf2/TGFβ1/Smad3 pathway. Eur J Pharmacol 2024; 963:176227. [PMID: 38072040 DOI: 10.1016/j.ejphar.2023.176227] [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: 04/13/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 01/07/2024]
Abstract
Skin fibrosis, the most obvious clinical manifestation of systemic sclerosis (SSc), has a high unmet need for treatment. Xanthohumol (Xn) has been shown to have beneficial effects on fibrotic diseases, but its efficacy in SSc remains unreported. This study aims to elucidate the effects and mechanisms of Xn on collagen synthesis in SSc skin fibroblasts (SScF). We found increased collagen production in SScF cultured in vitro, accompanied by dysregulated levels of oxidative stress. Cell experiments showed that Xn inhibited cell proliferation and promoted apoptosis. In addition, Xn was shown for the first time to upregulate reactive oxygen species (ROS) and nuclear factor erythroid 2-related factor 2 (Nrf2)levels in SScF, and when combined with the ROS scavenger N-acetylcysteine (NAC), Nrf2 expression was decreased. Importantly, we demonstrated that Xn significantly attenuated collagen synthesis by blocking the fibrotic classical transforming growth factor beta 1 (TGFβ1)/Smad3 pathway, which interestingly was upregulated when combined with the Nrf2 inhibitor 385. Taken together, Xn suppressed the TGFβ1/Smad3 pathway to ameliorate collagen overproduction by promoting ROS-induced oxidative stress damage and activating Nrf2, suggesting that Xn administration may be an emerging therapeutic strategy for skin fibrosis in SSc.
Collapse
Affiliation(s)
- Yu Xiao
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Zhongzhou Huang
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Yingyu Wang
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Yan Wang
- Central Lab, Huashan Hospital, Fudan University, Shanghai, China
| | - Ling Yu
- Shanghai TCM-Integrated Hospital, Shanghai University of TCM, Shanghai, China
| | - Ji Yang
- Division of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hejian Zou
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Weiguo Wan
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| | - Xue Yang
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| |
Collapse
|
41
|
Lv HW, Wang QL, Luo M, Zhu MD, Liang HM, Li WJ, Cai H, Zhou ZB, Wang H, Tong SQ, Li XN. Phytochemistry and pharmacology of natural prenylated flavonoids. Arch Pharm Res 2023; 46:207-272. [PMID: 37055613 PMCID: PMC10101826 DOI: 10.1007/s12272-023-01443-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 03/07/2023] [Indexed: 04/15/2023]
Abstract
Prenylated flavonoids are a special kind of flavonoid derivative possessing one or more prenyl groups in the parent nucleus of the flavonoid. The presence of the prenyl side chain enriched the structural diversity of flavonoids and increased their bioactivity and bioavailability. Prenylated flavonoids show a wide range of biological activities, such as anti-cancer, anti-inflammatory, neuroprotective, anti-diabetic, anti-obesity, cardioprotective effects, and anti-osteoclastogenic activities. In recent years, many compounds with significant activity have been discovered with the continuous excavation of the medicinal value of prenylated flavonoids, and have attracted the extensive attention of pharmacologists. This review summarizes recent progress on research into natural active prenylated flavonoids to promote new discoveries of their medicinal value.
Collapse
Affiliation(s)
- Hua-Wei Lv
- College of Pharmaceutical Science & Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products & Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, 310014, Hang zhou, P. R. China
| | - Qiao-Liang Wang
- College of Pharmaceutical Science & Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products & Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, 310014, Hang zhou, P. R. China
| | - Meng Luo
- College of Pharmaceutical Science & Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products & Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, 310014, Hang zhou, P. R. China
| | - Meng-Di Zhu
- Research Center of Analysis and Measurement, Zhejiang University of Technology University, 310014, Hang Zhou, P. R. China
| | - Hui-Min Liang
- College of Pharmaceutical Science & Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products & Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, 310014, Hang zhou, P. R. China
| | - Wen-Jing Li
- College of Pharmaceutical Science & Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products & Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, 310014, Hang zhou, P. R. China
| | - Hai Cai
- College of Pharmaceutical Science & Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products & Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, 310014, Hang zhou, P. R. China
| | - Zhong-Bo Zhou
- School of Pharmacy, Youjiang Medical University for Nationalities, 533000, Baise, P. R. China
| | - Hong Wang
- College of Pharmaceutical Science & Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products & Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, 310014, Hang zhou, P. R. China
| | - Sheng-Qiang Tong
- College of Pharmaceutical Science & Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products & Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, 310014, Hang zhou, P. R. China.
| | - Xing-Nuo Li
- College of Pharmaceutical Science & Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products & Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, 310014, Hang zhou, P. R. China.
| |
Collapse
|
42
|
Xanthohumol properties and strategies for extraction from hops and brewery residues: A review. Food Chem 2023; 404:134629. [DOI: 10.1016/j.foodchem.2022.134629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/16/2022] [Accepted: 10/13/2022] [Indexed: 11/22/2022]
|
43
|
Shu J, Hu L, Wu Y, Chen L, Huang K, Wang Z, Liang M. Daidzein suppresses TGF-β1-induced cardiac fibroblast activation via the TGF-β1/SMAD2/3 signaling pathway. Eur J Pharmacol 2022; 919:174805. [PMID: 35151651 DOI: 10.1016/j.ejphar.2022.174805] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 01/20/2022] [Accepted: 02/08/2022] [Indexed: 12/21/2022]
Abstract
Myocardial fibrosis is a concomitant bioprocess associated with many cardiovascular diseases (CVDs). Daidzein is an isoflavone that has been used for the treatment of CVDs. This study aimed to reveal its role in myocardial fibrosis. Our results indicate that daidzein had a nontoxic effect on cardiac fibroblasts and that TGF-β1 and TGFβRI levels were gradually decreased by daidzein in a dose-dependent manner. In the current study, we show that daidzein significantly inhibited TGF-β1-induced mRNA and protein expression of α-SMA, collagen I, and collagen III. Accordingly, immunofluorescence staining of α-SMA was performed. Daidzein also inhibited TGF-β1-induced cardiac fibroblast proliferation and migration. Mechanistically, daidzein inhibited the TGF-β/SMAD signaling pathway induced by TGF-β1 in cardiac fibroblasts. Additionally, daidzein ameliorated MI-induced cardiac dysfunction and cardiac fibrosis in vivo. Based on these findings, we conclude that daidzein reduces TGF-β1-induced cardiac fibroblast activation by partially regulating the TGF-β1/SMAD2/3 signaling pathway.
Collapse
Affiliation(s)
- Jiangcheng Shu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lizhi Hu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yichen Wu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long Chen
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaohui Wang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
44
|
Signaling pathways and targeted therapy for myocardial infarction. Signal Transduct Target Ther 2022; 7:78. [PMID: 35273164 PMCID: PMC8913803 DOI: 10.1038/s41392-022-00925-z] [Citation(s) in RCA: 374] [Impact Index Per Article: 124.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Although the treatment of myocardial infarction (MI) has improved considerably, it is still a worldwide disease with high morbidity and high mortality. Whilst there is still a long way to go for discovering ideal treatments, therapeutic strategies committed to cardioprotection and cardiac repair following cardiac ischemia are emerging. Evidence of pathological characteristics in MI illustrates cell signaling pathways that participate in the survival, proliferation, apoptosis, autophagy of cardiomyocytes, endothelial cells, fibroblasts, monocytes, and stem cells. These signaling pathways include the key players in inflammation response, e.g., NLRP3/caspase-1 and TLR4/MyD88/NF-κB; the crucial mediators in oxidative stress and apoptosis, for instance, Notch, Hippo/YAP, RhoA/ROCK, Nrf2/HO-1, and Sonic hedgehog; the controller of myocardial fibrosis such as TGF-β/SMADs and Wnt/β-catenin; and the main regulator of angiogenesis, PI3K/Akt, MAPK, JAK/STAT, Sonic hedgehog, etc. Since signaling pathways play an important role in administering the process of MI, aiming at targeting these aberrant signaling pathways and improving the pathological manifestations in MI is indispensable and promising. Hence, drug therapy, gene therapy, protein therapy, cell therapy, and exosome therapy have been emerging and are known as novel therapies. In this review, we summarize the therapeutic strategies for MI by regulating these associated pathways, which contribute to inhibiting cardiomyocytes death, attenuating inflammation, enhancing angiogenesis, etc. so as to repair and re-functionalize damaged hearts.
Collapse
|
45
|
Fan C, Wang Q, Chen Y, Ye T, Fan Y. Exosomes derived from bone mesenchymal stem cells attenuate myocardial fibrosis both in vivo and in vitro via autophagy activation: the key role of miR-199a-3p/mTOR pathway. Hum Cell 2022; 35:817-835. [PMID: 35190954 DOI: 10.1007/s13577-022-00680-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/25/2022] [Indexed: 12/11/2022]
Abstract
Autophagy suppression plays key a role during myocardial fibrosis (MF) progression. Exosomes from stem cells attenuate MF. The current study aimed to explain the antifibrosis effects of exosomes by focusing on microRNAs (miRs). MF was induced in rats using transverse aortic constriction (TAC) method and handled with exosomes from bone mesenchymal stem cells (BMSCs). The results of in vivo assays were verified with H9c2 cells. MiR expression profile was determined using microarray detection. The influence of miR-199a-3p modulation in vivo and in vitro on the antifibrosis effect of exosomes then was assessed. Exosomes attenuated MF by inhibiting inflammation, improving tissue structure, and inhibiting fibrosis-related indicators in TAC rats, and the effects were associated with autophagy activation. In H9c2 cells, exosomes suppressed cell viability, induced cell apoptosis, inhibited fibrosis-related indicators, while and the inhibition of autophagy by 3-MA would block the effect of exosomes. Based on the microarray detection, miR-199a-3p level was selected as therapeutic target. The inhibition of miR-199a-3p impaired the antifibrosis effects of exosomes on H9c2 cells, which was associated with autophagy inhibition. Collectively, exosomes from BMSCs exerted antifibrosis effects via the distant transfer of miR-199a-3p to heart tissues, which induced autophagy by inhibiting mTOR.
Collapse
Affiliation(s)
- Chenrong Fan
- Department of Cardiology, The First People's Hospital of Wenling, No. 333 Chuan'an South Road, Chengxi Street, Wenling, 317500, Zhejiang, China
| | - Qizeng Wang
- Department of Cardiology, The First People's Hospital of Wenling, No. 333 Chuan'an South Road, Chengxi Street, Wenling, 317500, Zhejiang, China
| | - Youjin Chen
- Department of Cardiology, The First People's Hospital of Wenling, No. 333 Chuan'an South Road, Chengxi Street, Wenling, 317500, Zhejiang, China
| | - Tingting Ye
- Department of Cardiology, The First People's Hospital of Wenling, No. 333 Chuan'an South Road, Chengxi Street, Wenling, 317500, Zhejiang, China
| | - Yuncao Fan
- Department of Cardiology, The First People's Hospital of Wenling, No. 333 Chuan'an South Road, Chengxi Street, Wenling, 317500, Zhejiang, China.
| |
Collapse
|
46
|
Jiménez-Uribe AP, Gómez-Sierra T, Aparicio-Trejo OE, Orozco-Ibarra M, Pedraza-Chaverri J. Backstage players of fibrosis: NOX4, mTOR, HDAC, and S1P; companions of TGF-β. Cell Signal 2021; 87:110123. [PMID: 34438016 DOI: 10.1016/j.cellsig.2021.110123] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/16/2022]
Abstract
The fibrotic process could be easily defined as a pathological excess of extracellular matrix deposition, leading to disruption of tissue architecture and eventually loss of function; however, this process involves a complex network of several signal transduction pathways. Virtually almost all organs could be affected by fibrosis, the most affected are the liver, lung, skin, kidney, heart, and eyes; in all of them, the transforming growth factor-beta (TGF-β) has a central role. The canonical and non-canonical signal pathways of TGF-β impact the fibrotic process at the cellular and molecular levels, inducing the epithelial-mesenchymal transition (EMT) and the induction of profibrotic gene expression with the consequent increase in proteins such as alpha-smooth actin (α-SMA), fibronectin, collagen, and other extracellular matrix proteins. Recently, it has been reported that some molecules that have not been typically associated with the fibrotic process, such as nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4), mammalian target of rapamycin (mTOR), histone deacetylases (HDAC), and sphingosine-1 phosphate (S1P); are critical in its development. In this review, we describe and discuss the role of these new players of fibrosis and the convergence with TGF-β signaling pathways, unveiling new insights into the panorama of fibrosis that could be useful for future therapeutic targets.
Collapse
Affiliation(s)
| | - Tania Gómez-Sierra
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico
| | - Omar Emiliano Aparicio-Trejo
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| | - Marisol Orozco-Ibarra
- Laboratorio de Neurobiología Molecular y Celular, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez, Av. Insurgentes Sur # 3877, La Fama, Alcaldía Tlalpan, CP 14269 Ciudad de México, Mexico
| | - José Pedraza-Chaverri
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico.
| |
Collapse
|
47
|
Girisa S, Saikia Q, Bordoloi D, Banik K, Monisha J, Daimary UD, Verma E, Ahn KS, Kunnumakkara AB. Xanthohumol from Hop: Hope for cancer prevention and treatment. IUBMB Life 2021; 73:1016-1044. [PMID: 34170599 DOI: 10.1002/iub.2522] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022]
Abstract
Cancer is a major public health concern due to high mortality and poor quality of life of patients. Despite the availability of advanced therapeutic interventions, most treatment modalities are not efficacious, very expensive, and cause several adverse side effects. The factors such as drug resistance, lack of specificity, and low efficacy of the cancer drugs necessitate developing alternative strategies for the prevention and treatment of this disease. Xanthohumol (XN), a prenylated chalcone present in Hop (Humulus lupulus), has been found to possess prominent activities against aging, diabetes, inflammation, microbial infection, and cancer. Thus, this manuscript thoroughly reviews the literature on the anti-cancer properties of XN and its various molecular targets. XN was found to exert its inhibitory effect on the growth and proliferation of cancer cells via modulation of multiple signaling pathways such as Akt, AMPK, ERK, IGFBP2, NF-κB, and STAT3, and also modulates various proteins such as Notch1, caspases, MMPs, Bcl-2, cyclin D1, oxidative stress markers, tumor-suppressor proteins, and miRNAs. Thus, these reports suggest that XN possesses enormous therapeutic potential against various cancers and could be potentially used as a multi-targeted anti-cancer agent with minimal adverse effects.
Collapse
Affiliation(s)
- Sosmitha Girisa
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Queen Saikia
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Devivasha Bordoloi
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Kishore Banik
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Javadi Monisha
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Uzini Devi Daimary
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Elika Verma
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, South Korea
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| |
Collapse
|