51
|
Naderi R, Seyhani A, Shirpoor A, Jafari A, Eyvani K. Effects of curcumin on cyclosporine A-induced oxidative stress, autophagy, and apoptosis in rat heart. Mol Biol Rep 2025; 52:310. [PMID: 40085292 DOI: 10.1007/s11033-025-10334-4] [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: 12/04/2024] [Accepted: 02/03/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Cyclosporine A (CsA) is a powerful immunosuppressant commonly used as a prophylaxis on transplant. However, it is associated with serious effects, including cardiotoxicity. Curcumin is a bioactive compound known for its anti-oxidative, anti-inflammatory, and anti-apoptotic effects. So, the present study investigated the possible protective effect of curcumin on CsA-induced heart injury in rats, focusing on oxidative stress, autophagy, and apoptosis. METHODS A total of 32 male Wistar rats were divided into control, sham (drug solvent), CsA (30 mg/kg BW), and curcumin + CsA (40 mg/kg BW, 30 mg/kg BW, respectively) groups. After 4 weeks of treatment, the heart was isolated for molecular assays. Immunoblot detected oxidative and autophagic proteins NOX4, hsp-70, beclin-1, and LC3II. The amount of 8-OHdG was measured by ELISA and heart apoptosis was examined by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining (TUNEL). RESULTS At the molecular levels, CSA increased the expression of NOX-4, beclin-1, LC3b, and oHdG in heart tissue. In addition, the amount of apoptosis increased in the heart tissue. However, curcumin treatment improved heart injury by significantly downregulating NOX4, LC3b, and decreasing 8-OHdG. Also, curcumin significantly reduced the rate of myocardial apoptosis. CONCLUSION To sum up, curcumin appears to protect against CsA-induced cardiotoxicity in rats by reducing oxidative activity, apoptosis, and regulating autophagy.
Collapse
Affiliation(s)
- Roya Naderi
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Alireza Shirpoor
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Adele Jafari
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Kimia Eyvani
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
52
|
Jaidee R, Jusakul A, Pocasap P, Kukongviriyapan V, Senggunprai L, Prawan A, Loilome W, Titapun A, Jareanrat A, Thanasukarn V, Khuntikeo N, Namwat N, Chamgramol Y, Thanee M, Wichian P, Hong JH, Guan P, Heng HL, Pairojkul C, Teh BT, Kongpetch S. Establishment and genomic profiling of cholangiocarcinoma cells with functional characterization. Sci Rep 2025; 15:8621. [PMID: 40074934 PMCID: PMC11904213 DOI: 10.1038/s41598-025-93192-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a highly lethal hepatobiliary malignancy, with prognosis is influenced by anatomical subtypes and etiological factors. This study successfully established three CCA cell lines: KKU-097, KKU-466, and KKU-610, from the primary tumors of patients in liver fluke-endemic areas. These cells represent the perihilar CCA (pCCA) and intrahepatic CCA (iCCA) subtypes. Comprehensive analyses, including histopathology, molecular profiling, biomarkers, cancer phenotype characterization, and drug sensitivity testing with standard chemotherapeutics, were conducted. Whole-exome sequencing was performed to explore genetic alterations. All three cell lines exhibited adherent growth with an epithelial morphology and positive expression of the bile duct epithelial markers CK-7 and CK-19. Cytogenetic analysis revealed highly complex hypertriploid karyotypes with multiple chromosomal aberrations. Among the cell lines, KKU-610 demonstrated higher growth and invasion rates, whereas KKU-466 and KKU-097 cells exhibited less aggressive phenotypes. Drug sensitivity testing demonstrated relative resistance to gemcitabine as a monotherapy and in combination with cisplatin in all three cells. Genomic profiling identified targetable mutations, highlighting these new cell lines as valuable models for investigating the pathogenesis of CCA and evaluating therapeutic strategies.
Collapse
Affiliation(s)
- Rattanaporn Jaidee
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Apinya Jusakul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Piman Pocasap
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Veerapol Kukongviriyapan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Laddawan Senggunprai
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Auemduan Prawan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharin Loilome
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Attapol Titapun
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Apiwat Jareanrat
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Vasin Thanasukarn
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Natcha Khuntikeo
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nisana Namwat
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Yaovalux Chamgramol
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Malinee Thanee
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Phongsathorn Wichian
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Jing Han Hong
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Peiyong Guan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hong Lee Heng
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Center Singapore, Singapore, Singapore
| | - Chawalit Pairojkul
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Bin Tean Teh
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Center Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Sarinya Kongpetch
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
53
|
Kumar H, Dhalaria R, Kimta N, Guleria S, Upadhyay NK, Nepovimova E, Dhanjal DS, Sethi N, Manickam S. Curcumin: A Potential Detoxifier Against Chemical and Natural Toxicants. Phytother Res 2025; 39:1494-1530. [PMID: 39853860 DOI: 10.1002/ptr.8442] [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: 10/08/2024] [Revised: 01/05/2025] [Accepted: 01/11/2025] [Indexed: 01/26/2025]
Abstract
The human body gets exposed to a variety of toxins intentionally or unintentionally on a regular basis from sources such as air, water, food, and soil. Certain toxins can be synthetic, while some are biological. The toxins affect the various parts of the body by activating numerous pro-inflammatory markers, like oxidative stresses, that tend to disturb the normal function of the organs ultimately. Nowadays, people use different types of herbal treatments, viz., herbal drinks that contain different spices for detoxification of their bodies. One such example is turmeric, the most commonly available spice in the kitchen and used across all kinds of households. Turmeric contains curcumin, which is a natural polyphenol. Curcumin is a medicinal compound with different biological activities, such as antioxidant, antineoplastic, anti-inflammatory, and antibacterial. Hence, this review gives a comprehensive insight into the promising potential of curcumin in the detoxification of heavy metals, carbon tetrachloride, drugs, alcohol, acrylamide, mycotoxins, nicotine, and plastics. The review encompasses diverse animal-based studies portraying curcumin's role in nullifying the different toxic effects in various organs of the body (especially the liver, kidney, testicles, and brain) by enhancing defensive signaling pathways, improving antioxidant enzyme levels, inhibiting pro-inflammatory markers activities and so on. Furthermore, this review also argues over curcumin's safety assessment for its utilization as a detoxifying agent.
Collapse
Affiliation(s)
- Harsh Kumar
- Centre of Advanced Technologies, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Rajni Dhalaria
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Neetika Kimta
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Shivani Guleria
- Department of Biotechnology, TIFAC-Centre of Relevance and Excellence in Agro and Industrial Biotechnology (CORE), Thapar Institute of Engineering and Technology, Patiala, India
| | | | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
- Centre for Advanced Innovation Technologies, VSB-Technical University of Ostrava, Ostrava-Poruba, Czech Republic
| | - Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
| | - Nidhi Sethi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Sivakumar Manickam
- Chemical and Energy Engineering, Faculty of Engineering, Universiti Teknologi Brunei, Bandar Seri Begawan, Brunei
| |
Collapse
|
54
|
Suzuki K, Watanabe N, Torii S, Arakawa S, Ochi K, Tsuchiya S, Yamada K, Kawamura Y, Ota S, Komatsu N, Shimizu S, Ando M, Takaku T. BCR::ABL1-induced mitochondrial morphological alterations as a potential clinical biomarker in chronic myeloid leukemia. Cancer Sci 2025; 116:673-689. [PMID: 39652455 PMCID: PMC11875769 DOI: 10.1111/cas.16424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 03/05/2025] Open
Abstract
The BCR::ABL1 oncogene plays a crucial role in the development of chronic myeloid leukemia (CML). Previous studies have investigated the involvement of mitochondrial dynamics in various cancers, revealing potential therapeutic strategies. However, the impact of BCR::ABL1 on mitochondrial dynamics remains unclear. In this study, we demonstrated that BCR::ABL1 is sufficient to induce excessive mitochondrial fragmentation by activating dynamin-related protein (DRP)1 through the mitogen-activated protein kinase (MAPK) pathway. Leukocytes obtained from patients with CML and the BCR::ABL1-positive cell lines exhibited increased mitochondrial fragmentation compared to leukocytes obtained from healthy donors and BCR::ABL1-negative cells. Furthermore, the analysis of BCR::ABL1-transduced cells showed increased phosphorylation of DRP1 at serine 616 and extracellular signal-regulated kinase (ERK) 1/2. Moreover, the inhibition of DRP1 and upstream mitogen-activated extracellular signal-regulated kinase (MEK) 1/2 suppressed mitochondrial fragmentation. Strikingly, DRP1 inhibition effectively reduced the viability of BCR::ABL1-positive cells and induced necrotic cell death. Additionally, a label-free artificial intelligence-driven flow cytometry successfully identified not only the BCR::ABL1-transduced cells but also peripheral leukocytes from CML patients by assessing mitochondrial morphological alterations. These findings suggested the crucial role of BCR::ABL1-induced mitochondrial fragmentation in driving BCR::ABL1-positive cell proliferation, and the potential use of mitochondrial morphological alterations as a clinical biomarker for the label-free detection of CML cells.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Fusion Proteins, bcr-abl/metabolism
- Fusion Proteins, bcr-abl/genetics
- Mitochondria/metabolism
- Mitochondria/pathology
- Mitochondria/genetics
- Dynamins/metabolism
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Phosphorylation
- MAP Kinase Signaling System
- Mitochondrial Dynamics/genetics
Collapse
Affiliation(s)
- Kohjin Suzuki
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
- System Technologies Laboratory, Sysmex CorporationKobeJapan
| | - Naoki Watanabe
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Satoru Torii
- Department of Pathological Cell BiologyAdvanced Research Initiative, Institute of Science TokyoTokyoJapan
| | - Satoko Arakawa
- Department of Pathological Cell BiologyAdvanced Research Initiative, Institute of Science TokyoTokyoJapan
| | - Kiyosumi Ochi
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
- Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Shun Tsuchiya
- Department of HematologyJuntendo University Nerima HospitalTokyoJapan
| | | | | | - Sadao Ota
- ThinkCyteK.K.TokyoJapan
- Research Center for Advanced Science and TechnologyThe University of TokyoTokyoJapan
| | - Norio Komatsu
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Shigeomi Shimizu
- Department of Pathological Cell BiologyAdvanced Research Initiative, Institute of Science TokyoTokyoJapan
| | - Miki Ando
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Tomoiku Takaku
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
- Department of HematologySaitama Medical UniversitySaitamaJapan
| |
Collapse
|
55
|
He J, Liao JH. Potential Role of Hesperidin in Improving Experimental Pulmonary Arterial Hypertension in Rats via Modulation of the NF-κB Pathway. Chem Biol Drug Des 2025; 105:e70068. [PMID: 40110966 DOI: 10.1111/cbdd.70068] [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: 04/29/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 03/22/2025]
Abstract
This study was designed to evaluate the therapeutic effects of hesperidin, an anti-inflammatory compound, on pulmonary arterial hypertension (PAH). A PAH rat model was established using monocrotaline (MCT, 60 mg/kg). Next, the experimental animals were assigned into the following four groups (n = 6 per group): Control group, MCT group, MCT + H20 group (20 mg/kg hesperidin), and MCT + H40 group (40 mg/kg hesperidin). According to the experimental outcomes, the PAH rat model was built successfully. In PAH animals, hesperidin significantly reduced right ventricular systolic pressure, Fulton index, and mean pulmonary arterial pressure. Concurrently, it improved pulmonary artery velocity-time integral and acceleration time, as well as alleviated pulmonary artery and right ventricular remodeling. On a molecular level, hesperidin inhibited the expression of vascular endothelial-cadherin, alpha-smooth muscle actin, matrix metalloproteinase-9, and transforming growth factor beta. Also, hesperidin downregulated pro-inflammatory cytokines such as interleukin (IL)-6, IL-1β, IL-18, chemokine C-C motif ligand 2, and C-X-C motif chemokine ligand 1 levels, and reduced the number of CD68+ cells in tissue samples. Further analysis revealed that hesperidin could inhibit the activation of p-IκB-α and p-p65 in samples induced by MCT. Collectively, these findings suggest that hesperidin may inhibit inflammation through the NF-κB pathway, thereby improving experimental PAH in rats induced by MCT.
Collapse
Affiliation(s)
- Jun He
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | | |
Collapse
|
56
|
Gao XQ, Li HL, Wang M, Yang CT, Su R, Shao LH. Kaempferol inhibited invasion and metastasis of gastric cancer cells by targeting AKT/GSK3β pathway based on network pharmacology and molecular docking. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025; 27:421-441. [PMID: 39132822 DOI: 10.1080/10286020.2024.2387756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
This study aims to explore the mechanisms of the inhibitory effect of kaempferol on the invasion and metastasis of gastric cancer (GC) cells through network pharmacology prediction and experimental verification. It identifies core targets via PPI network analysis and finds that kaempferol binds to these targets well. In vitro experiments showed that kaempferol could inhibit the proliferation, colony formation, migration and invasion of GC cells. Western blotting indicated kaempferol may reduce AKT and GSK3β phosphorylation, leading to lower expression of invasion-related genes SRC, MMP9, CXCR4, KDR, and MMP2. Overall, kaempferol may prevent migration and invasion of GC cells via the AKT/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Xia-Qing Gao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Hai-Long Li
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Department of Geriatrics, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Meng Wang
- Department of Geriatrics, Affiliated Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, China
- Research Center of Traditional Chinese Medicine, Gansu Province, Lanzhou 730000, China
| | - Chun-Ting Yang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Rong Su
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Li-Hua Shao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
| |
Collapse
|
57
|
Putatunda V, Jusakul A, Roberts L, Wang XW. Genetic, Epigenetic, and Microenvironmental Drivers of Cholangiocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:362-377. [PMID: 39532242 PMCID: PMC11841490 DOI: 10.1016/j.ajpath.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Cholangiocarcinoma (CCA) is an aggressive and heterogeneous malignancy of the biliary tree that carries a poor prognosis. Multiple features at the genetic, epigenetic, and microenvironmental levels have been identified to better characterize CCA carcinogenesis. Genetic alterations, such as mutations in IDH1/2, BAP1, ARID1A, and FGFR2, play significant roles in CCA pathogenesis, with variations across different subtypes, races/ethnicities, and causes. Epigenetic dysregulation, characterized by DNA methylation and histone modifications, further contributes to the complexity of CCA, influencing gene expression and tumor behavior. Furthermore, CCA cells exchange autocrine and paracrine signals with other cancer cells and the infiltrating cell types that populate the microenvironment, including cancer-associated fibroblasts and tumor-associated macrophages, further contributing to an immunosuppressive niche that supports tumorigenesis. This review explores the multifaceted genetic, epigenetic, and microenvironmental drivers of CCA. Understanding these diverse mechanisms is essential for characterizing the complex pathways of CCA carcinogenesis and developing targeted therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Vijay Putatunda
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland; Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| | - Apinya Jusakul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand; Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Lewis Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Xin Wei Wang
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland; Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
58
|
Wei X, He Y, Yu Y, Tang S, Liu R, Guo J, Jiang Q, Zhi X, Wang X, Meng D. The Multifaceted Roles of BACH1 in Disease: Implications for Biological Functions and Therapeutic Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412850. [PMID: 39887888 PMCID: PMC11905017 DOI: 10.1002/advs.202412850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/22/2024] [Indexed: 02/01/2025]
Abstract
BTB domain and CNC homolog 1 (BACH1) belongs to the family of basic leucine zipper proteins and is expressed in most mammalian tissues. It can regulate its own expression and play a role in transcriptionally activating or inhibiting downstream target genes. It has a crucial role in various biological processes, such as oxidative stress, cell cycle, heme homeostasis, and immune regulation. Recent research highlights BACH1's significant regulatory roles in a series of conditions, including stem cell pluripotency maintenance and differentiation, growth, senescence, and apoptosis. BACH1 is closely associated with cardiovascular diseases and contributes to angiogenesis, atherosclerosis, restenosis, pathological cardiac hypertrophy, myocardial infarction, and ischemia/reperfusion (I/R) injury. BACH1 promotes tumor cell proliferation and metastasis by altering tumor metabolism and the epithelial-mesenchymal transition phenotype. Moreover, BACH1 appears to show an adverse role in diseases such as neurodegenerative diseases, gastrointestinal disorders, leukemia, pulmonary fibrosis, and skin diseases. Inhibiting BACH1 may be beneficial for treating these diseases. This review summarizes the role of BACH1 and its regulatory mechanism in different cell types and diseases, proposing that precise targeted intervention of BACH1 may provide new strategies for human disease prevention and treatment.
Collapse
Affiliation(s)
- Xiangxiang Wei
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Yunquan He
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Yueyang Yu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Sichong Tang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Ruiwen Liu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Jieyu Guo
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Qingjun Jiang
- Department of Vascular & Endovascular SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Xiuling Zhi
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Xinhong Wang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Dan Meng
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| |
Collapse
|
59
|
Macias RIR, Kanzaki H, Berasain C, Avila MA, Marin JJG, Hoshida Y. The Search for Risk, Diagnostic, and Prognostic Biomarkers of Cholangiocarcinoma and Their Biological and Clinicopathologic Significance. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:422-436. [PMID: 39103092 PMCID: PMC11841489 DOI: 10.1016/j.ajpath.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/01/2024] [Accepted: 06/20/2024] [Indexed: 08/07/2024]
Abstract
Cholangiocarcinomas (CCAs) are a heterogeneous group of malignant tumors that originate from the biliary tract. They are usually diagnosed in advanced stages, leading to a poor prognosis for affected patients. As CCA often arises as a sporadic cancer in individuals lacking specific risk factors or with heterogeneous backgrounds, and there are no defined high-risk groups, the implementation of effective surveillance programs for CCA is problematic. The identification and validation of new biomarkers useful for risk stratification, diagnosis, prognosis, and prediction of treatment response remains an unmet need for patients with CCA, even though numerous studies have been conducted lately to try to discover and validate CCA biomarkers. In this review, we overview the available information about the different types of biomarkers that have been investigated in recent years using minimally invasive biospecimens (blood, serum/plasma, bile, and urine) and their potential usefulness in diagnosis, prognosis, and risk stratification. It is widely accepted that early detection of CCA will impact patients' outcomes, by improving survival rates, quality of life, and the possibility of less invasive and/or curative treatments; however, challenges to its translation and clinical application for patients with CCA need to be resolved.
Collapse
Affiliation(s)
- Rocio I R Macias
- Experimental Hepatology and Drug Targeting Group, University of Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases, Carlos III National Institute of Health, Madrid, Spain.
| | - Hiroaki Kanzaki
- Division of Digestive and Liver Diseases, Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Carmen Berasain
- Center for the Study of Liver and Gastrointestinal Diseases, Carlos III National Institute of Health, Madrid, Spain; Hepatology Laboratory, Solid Tumors Program, Center for Applied Medical Research, Cancer Center University of Navarra, Pamplona, Spain
| | - Matias A Avila
- Center for the Study of Liver and Gastrointestinal Diseases, Carlos III National Institute of Health, Madrid, Spain; Hepatology Laboratory, Solid Tumors Program, Center for Applied Medical Research, Cancer Center University of Navarra, Pamplona, Spain
| | - Jose J G Marin
- Experimental Hepatology and Drug Targeting Group, University of Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Center for the Study of Liver and Gastrointestinal Diseases, Carlos III National Institute of Health, Madrid, Spain
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
60
|
Zhang RL, Wang WM, Li JQ, Li RW, Zhang J, Wu Y, Liu Y. The role of miR-155 in cardiovascular diseases: Potential diagnostic and therapeutic targets. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2025; 24:200355. [PMID: 39760132 PMCID: PMC11699627 DOI: 10.1016/j.ijcrp.2024.200355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025]
Abstract
Cardiovascular diseases (CVDs), such as atherosclerotic cardiovascular diseases, heart failure (HF), and acute coronary syndrome, represent a significant threat to global health and impose considerable socioeconomic burdens. The intricate pathogenesis of CVD involves various regulatory mechanisms, among which microRNAs (miRNAs) have emerged as critical posttranscriptional regulators. In particular, miR-155 has demonstrated differential expression patterns across a spectrum of CVD and is implicated in the etiology and progression of arterial disorders. This systematic review synthesizes current evidence on the multifaceted roles of miR-155 in the modulation of genes and pathological processes associated with CVD. We delineate the potential of miR-155 as a diagnostic biomarker and therapeutic target, highlighting its significant regulatory influence on conditions such as atherosclerosis, aneurysm, hypertension, HF, myocardial hypertrophy, and oxidative stress. Our analysis underscores the transformative potential of miR-155 as a target for intervention in cardiovascular medicine, warranting further investigation into its clinical applicability.
Collapse
Affiliation(s)
- Rui-Lin Zhang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Wei-Ming Wang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, 646000, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ji-Qiang Li
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Run-Wen Li
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Jie Zhang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, 646000, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yong Liu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, 646000, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
61
|
Ellis H, Braconi C, Valle JW, Bardeesy N. Cholangiocarcinoma Targeted Therapies: Mechanisms of Action and Resistance. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:437-452. [PMID: 39730074 PMCID: PMC11841491 DOI: 10.1016/j.ajpath.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/29/2024]
Abstract
Cholangiocarcinoma is an aggressive bile duct malignancy with heterogeneous genomic features. Although most patients receive standard-of-care chemotherapy/immunotherapy, genomic changes that can be targeted with established or emerging therapeutics are common. Accordingly, precision medicine strategies are transforming the next-line treatment for patient subsets. Hotspot IDH1 mutations and activating fibroblast growth factor receptor 2 fusions occur frequently, and small-molecule inhibitors against these alterations are US Food and Drug Administration approved. Translational and basic science studies have elucidated the mechanisms of response and resistance in cholangiocarcinoma, providing insights into these targets that extend to other cancers. Additional US Food and Drug Administration-approved and National Comprehensive Cancer Network guideline-recommended treatments for recurrent genomic changes include BRAF inhibition (BRAF-V600E) and trastumazab deruxtecan (human epidermal growth factor receptor 2 amplification). Furthermore, ongoing clinical trials show promising results with KRAS inhibition (KRAS-codon 12 mutations), PRTM5 inhibition, alone or with methylthioadenosine inhibition (5-methylthioadenosine phosphorylase deletion), and murine double minute 2 inhibition (murine double minute 2 amplification). Despite these advances, the rate, depth, and duration of response to each treatment need improvement. Moreover, many patients do not have currently targetable genotypes. This review examines the clinical efficacy and mechanisms of resistance associated with these treatments, as well as insights into the molecular and biological effects of pathway activation and inhibition, based on study of patient samples and preclinical models. It also explores strategies to overcome resistance and possible precision medicine approaches for additional patient subsets.
Collapse
Affiliation(s)
- Haley Ellis
- Massachusetts General Hospital Cancer Center, Krantz Family Center for Cancer Research, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Chiara Braconi
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom; Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Juan W Valle
- Cholangiocarcinoma Foundation, Herriman, Utah; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Krantz Family Center for Cancer Research, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
62
|
Sosa-García C, Sánchez-Pacheco UA, Tavira-Montalvan CA, Meneses-Acosta A. Expression of UCOE and HSP27 Molecular Elements to Improve the Stable Protein Production on HEK293 Cells. BIOMED RESEARCH INTERNATIONAL 2025; 2025:5556353. [PMID: 40041570 PMCID: PMC11879590 DOI: 10.1155/bmri/5556353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/11/2025] [Indexed: 03/06/2025]
Abstract
Recombinant proteins represent one of the greatest achievements of modern pharmaceutical biotechnology, as they are increasingly used across nearly all branches of medicine to treat a wide range of conditions. In response to this demand, various cell engineering approaches have been developed to improve their expression. Some of these approaches involve the use of genetic elements that prevent the silencing of the gene of interest, as well as the generation of resistant cell lines to inhibit or avoid programmed cell death (PCD). This research focuses on analyzing the effects of overexpression of UCOE elements and the HSP27 protein, both individually and together, on the production of human rIFNγ in HEK293 cells. Our results show that 4-Kb UCOE elements have no effect on protein production in HEK293 cells, while overexpression of HSP27 prolongs the stationary phase during growth kinetics. The Qp of rIFNγ is 96-fold higher in clones containing the HSP27/UCOE combination compared to the clone containing only UCOE elements or to the control HEK293 cells. These results correlate with the MCP analyses, which showed that overexpression of HSP27 decreased the expression of Bax, caspase 3, cytochrome C, Beclin, and LC3II mRNA. Finally, this study suggests the potential utility of a cell engineering approach based on the overexpression of the human HSP27 protein for enhancing the production of recombinant viruses and proteins in HEK293 cells.
Collapse
Affiliation(s)
- Concepción Sosa-García
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmacy, Autonomous University of the State of Morelos, Cuernavaca, Morelos, Mexico
| | - Uriel Abdallah Sánchez-Pacheco
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmacy, Autonomous University of the State of Morelos, Cuernavaca, Morelos, Mexico
| | - Carlos Alberto Tavira-Montalvan
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmacy, Autonomous University of the State of Morelos, Cuernavaca, Morelos, Mexico
| | - Angélica Meneses-Acosta
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmacy, Autonomous University of the State of Morelos, Cuernavaca, Morelos, Mexico
| |
Collapse
|
63
|
Khamseekaew J, Duangjinda M, Maneesai P, Labjit C, Rattanakanokchai S, Rongpan S, Pakdeechote P, Potue P. Cannabis sativa L. Leaf Oil Displays Cardiovascular Protective Effects in Hypertensive Rats. Int J Mol Sci 2025; 26:1897. [PMID: 40076524 PMCID: PMC11899380 DOI: 10.3390/ijms26051897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Hemp (Cannabis sativa L.) leaf oil (HLO) contains several bioactive compounds such as phenolics, flavonoids, and quercetin. However, the effects of HLO on hypertensive conditions have not yet been investigated. This study investigated the cardiovascular protective effects of HLO in a nitric oxide (NO) synthase inhibitor-induced hypertensive rat model. Five weeks of HLO administration significantly prevented blood pressure elevation, improved cardiac function, and mitigated cardiac hypertrophy. Furthermore, HLO ameliorated vascular dysfunction by reducing sympathetic nerve stimulation-induced vasoconstriction, increasing endothelium-dependent vasorelaxation, as well as decreasing vascular wall thickness and vascular smooth muscle cell proliferation. HLO inhibited renin-angiotensin system (RAS) activation and downregulated angiotensin II type 1 (AT1) receptor and NADPH oxidase expression. Additionally, HLO normalized the circulating NO metabolites, decreased oxidative stress, and enhanced antioxidant status. These findings suggest that HLO protects against cardiovascular dysfunction and preserves its morphology. The mechanism of action might involve the suppression of RAS overactivity and oxidative stress through the Ang II/AT1 receptor/NOX2 pathway in NO-deficient hypertension.
Collapse
Affiliation(s)
- Juthamas Khamseekaew
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (J.K.); (P.M.); (S.R.); (P.P.)
| | - Monchai Duangjinda
- Department of Animal Science, Faculty of Agriculture, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Putcharawipa Maneesai
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (J.K.); (P.M.); (S.R.); (P.P.)
| | - Chanon Labjit
- Department of Horticulture, Faculty of Agriculture, Khon Kaen University, Khon Kaen 40002, Thailand;
| | | | - Sudarat Rongpan
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (J.K.); (P.M.); (S.R.); (P.P.)
- Department of Physiology, Faculty of Medicine, Princess of Naradhiwas University, Narathiwat 96000, Thailand
| | - Poungrat Pakdeechote
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (J.K.); (P.M.); (S.R.); (P.P.)
| | - Prapassorn Potue
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (J.K.); (P.M.); (S.R.); (P.P.)
| |
Collapse
|
64
|
Yan CY, Ju F, Chen YH, Mu YT, Hu YJ, Guo DL, Deng Y, Kuang QX. Two undescribed benzofuran derivatives from chaetomium elatum. Nat Prod Res 2025:1-5. [PMID: 39985178 DOI: 10.1080/14786419.2025.2469306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
Two undescribed benzofuran derivatives (1-2) were isolated from Chaetomium elatum using silica gel, Sephadex LH-20 column chromatography, and preparative high-performance liquid chromatography (HPLC). The structures of compound 1-2 were characterised by HRESIMS, NMR, HMBC, NOESY spectral analyses, and ECD calculations. Compounds 1-2 demonstrated the ability to mitigate the inflammatory response triggered by lipopolysaccharide (LPS) in RAW264.7 macrophages, as evidenced by RT-PCR assay results showing suppression of key inflammatory factors at the concentration of 30 μM. These findings suggest that 1-2 possess promising anti-inflammatory properties.
Collapse
Affiliation(s)
- Chen-Yi Yan
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine Chengdu, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feng Ju
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Yu-Hui Chen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Yu-Ting Mu
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yun-Jie Hu
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Da-Le Guo
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yun Deng
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qi-Xuan Kuang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine Chengdu, Chengdu, China
| |
Collapse
|
65
|
Nazeam JA, Black I, Mulamoottil VA, Selim NM, El Shiekh RA, Abu-Elfotuh K, Hamdan AME, Gowifel AMH, Hafez SM, Mohamed EK, Atwa AM, El Hefnawy HM, Azadi P. Okra seed polysaccharides mitigate neuroinflammation and cognitive impairment via modulation of Nrf2/HO-1, HMGB1/RAGE/TLR4/NF-κB, NLRP3/Caspase-1, JAK-2/STAT-3, AMPK/SIRT1/m-TOR, PI3K/AKT/CREB/BDNF/TrkB and PERK/CHOP/Bcl-2 axes. Int Immunopharmacol 2025; 148:114110. [PMID: 39862637 DOI: 10.1016/j.intimp.2025.114110] [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: 08/04/2024] [Revised: 11/23/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Global healthcare systems are under tremendous strain due to the increasing prevalence of neurodegenerative disorders. Growing data suggested that overconsumption of high-fat/high-carbohydrates diet (HFHCD) is associated with enhanced incidence of metabolic alterations, neurodegeneration, and cognitive dysfunction. Functional foods have gained prominence in curbing metabolic and neurological deficits. Consequently, this study endeavored to explore effects of purified Okra seed polysaccharides (OP) (Abelmoschus esculentus (L.) Moench) against HFHCD-induced metabolic alterations and cognitive dysfunction, with elucidating underlying contributed mechanistic pathways. OP hydrolysate was analyzed using GC-MS analysis. The biological study encompassed two phases, the first phase I (model establishment phase), for 3 months, involved a control group, fed standard diet, and HFHCD group. The second phase (phase II) where HFHCD fed rats were re-divided into 3 equal subgroups, 1st subgroup received HFHCD, whereas second and third subgroups received OP, 200 or 400 mg/kg/day, respectively, for 28 days. GC-MS characterized OP as an arabinogalactouranan and revealed the monosaccharide composition as galacturonic acid: arabinose: glucose: galactose: rhamnose: xylose in ratio of 28.2: 23.3: 11.5: 4.2: 3.5: 2.0. The findings demonstrated that OP dose-dependently mitigated HFHCD-induced rise in body weights, lipid profiles, levels of blood glucose and disruption in behavioral outcomes, neurotransmitters, together with histopathological alterations in brain. Moreover, OP dose-dependently improved redox, neuroinflammatory, endoplasmic reticulum (ER) stress, autophagic and apoptotic biomarkers. OP can be regarded as promising functional food candidate to hamper HFHCD-induced metabolic alterations and cognitive deficit, via enhancing Nrf2/HO-1, AMPK/SIRT1 and PI3K/AKT/CREB axes, long with dampening of HMGB1/RAGE/TLR4, NLRP3/Caspase-1, JAK-2/STAT-3 and PERK/CHOP axes.
Collapse
Affiliation(s)
- Jilan A Nazeam
- Department of Pharmacognosy, Faculty of Pharmacy, October 6 University, Giza, Egypt; Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.
| | - Ian Black
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.
| | | | - Nabil M Selim
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Giza 11562, Egypt.
| | - Riham A El Shiekh
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Giza 11562, Egypt.
| | - Karema Abu-Elfotuh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt; Al-Ayen Iraqi University, Thi-Qar 64001, Iraq.
| | - Ahmed M E Hamdan
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; Prince Fahad bin Sultan Chair for Biomedical Research, University of Tabuk, Saudi Arabia.
| | - Ayah M H Gowifel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt.
| | - Shaimaa M Hafez
- Department of Anatomy and Embryology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt.
| | - Ehsan K Mohamed
- Biochemistry Department, Egyptian Drug Authority (EDA), Formerly National Organization of Drug Control and Research (NODCAR), Giza, Egypt
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo-Suez Road, Cairo 11829, Egypt; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Ayen Iraqi University, Thi-Qar, 64001, Iraq.
| | | | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.
| |
Collapse
|
66
|
Wu JS, Chu PY, Hsu WY, Chuang TH, Yu YC, Pan YC, Lin YT, Tang CH, Lee CL, Wu YC. Fractionation and identification of ocular protective compounds from kochiae fructus against oxidative damage in retinal pigment epithelium cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119328. [PMID: 39793774 DOI: 10.1016/j.jep.2025.119328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/26/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kochiae Fructus, the ripe fruit of Kochia scoparia, is a traditional Chinese medicine commonly used to treat eye discomforts and vision problems. Although Kochiae Fructus is mentioned in many classical Chinese medical texts, its protective effects and the roles of its active phytochemicals in eye treatment still lack scientific exploration. AIM OF THE STUDY This study aimed to clarify the protective effects and identify the active fractions and compounds of Kochiae Fructus against oxidative stress-induced retinal pigment epithelium (RPE) cell death. MATERIALS AND METHODS Liquid-liquid partitioning was employed to prepare active fractions. Silica gel, RP-18, and Sephadex™ LH-20 gels were used as stationary phases to purify the compounds through column chromatography. Cell models were established by treating ARPE-19 (RPE cell line) with hydrogen peroxide (H₂O₂) and tert-butyl hydroperoxide (TBHP), respectively. RESULTS The methanol aqueous fraction from ethyl acetate-soluble extract (KSEM) alleviated oxidant-induced RPE cell death in a dose-dependent manner and activated the Nrf2/HO-1 pathway, a critical defense mechanism against oxidative damage. The isolated flavonols in fraction KSEM, hyperoside, quercetin, and kaempferol, reduced oxidant-induced cell mortality, with quercetin showing the strongest effect. Moreover, combination treatment revealed that hyperoside and kaempferol synergistically enhanced the protective effects of quercetin, making the combined treatment more efficient than quercetin alone. CONCLUSIONS The protective effects of Kochiae Fructus against oxidative damage in RPE cell was validated with the KSEM fraction. The compound quercetin was identified as the important contributor. However, the synergistic effect of the hyperoside, quercetin, and kaempferol mixture may play a dominant role in the protective action of Kochiae Fructus. Overall, these findings highlight the potential of Kochiae Fructus and its flavonol mixture as a basis for developing treatments targeting ocular diseases associated with oxidative damage in retina.
Collapse
Affiliation(s)
- Jian-Sheng Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 0402, Taiwan; Department of Ophthalmology, Changhua Christian Hospital, Changhua, 50006, Taiwan.
| | - Pei-Yi Chu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan; Master Program of Pharmaceutical Manufacture, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan.
| | - Wei-Yu Hsu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan.
| | - Ta-Hsien Chuang
- School of Pharmacy, China Medical University, Taichung, 406040, Taiwan.
| | - Ying-Chun Yu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan.
| | - Yi-Cheng Pan
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan.
| | - Yi-Ting Lin
- Department of Cosmeceutics, China Medical University, Taichung, 40604, Taiwan.
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 0402, Taiwan; Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, 40402, Taiwan.
| | - Chia-Lin Lee
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan; Department of Cosmeceutics, China Medical University, Taichung, 40604, Taiwan.
| | - Yang-Chang Wu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 40447, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan; Master Program of Pharmaceutical Manufacture, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, 41354, Taiwan.
| |
Collapse
|
67
|
Guerra-Ojeda S, Suarez A, Belmonte B, Marchio P, Genovés P, Arias-Mutis OJ, Aldasoro M, Vila JM, Serna E, Mauricio MD. Sodium valproate reverses aortic hypercontractility in acute myocardial infarction in rabbits. Eur J Pharmacol 2025; 988:177216. [PMID: 39706463 DOI: 10.1016/j.ejphar.2024.177216] [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: 09/02/2024] [Revised: 12/01/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Sympathetic nervous system (SNS), endothelin 1 (ET-1) and angiotensin II (Ang II) are involved in the pathophysiology of acute myocardial infarction (AMI). Valproic acid (VPA) is under study for the treatment against AMI due to its beneficial cardiac effects. However, the vascular effects of VPA on the activation of the SNS, ET-1 and Ang II after AMI are not fully studied. In our study, we used aorta from New Zealand White rabbits without AMI, with AMI and AMI treated with VPA (500 mg/kg/day). AMI was induced by occluding the left circumflex coronary artery for 1 h, followed by reperfusion. After 5 weeks, we studied the ex vivo vascular reactivity in organ bath and measured protein expression by Western blot. Our findings indicated that AMI increased vasoconstriction to exogenous and endogenous NE and ET-1, which was reversed by VPA eliciting upregulation of α2-adrenergic and ETB receptors and downregulating ETA receptors. Although no changes in the vascular response to Ang II were observed in AMI or VPA-treated rabbits, an increase in Ang II type 2 receptor expression was found in VPA-treated rabbits. We conclude that VPA could be considered a vascular protector by modulating SNS, ET-1 and Ang II in the aorta, which together with its cardioprotective effect would make it a promising candidate for the treatment of AMI.
Collapse
Affiliation(s)
- S Guerra-Ojeda
- Department of Physiology, School of Medicine, University of Valencia, Spain; Institute of Health Research INCLIVA, Valencia, Spain
| | - A Suarez
- Department of Physiology, School of Medicine, University of Valencia, Spain; Institute of Health Research INCLIVA, Valencia, Spain
| | - B Belmonte
- Department of Physiology, School of Medicine, University of Valencia, Spain; Institute of Health Research INCLIVA, Valencia, Spain
| | - P Marchio
- Department of Physiology, School of Medicine, University of Valencia, Spain; Institute of Health Research INCLIVA, Valencia, Spain
| | - P Genovés
- Department of Physiology, School of Medicine, University of Valencia, Spain; Institute of Health Research INCLIVA, Valencia, Spain; Center for Biomedical Research Network on Cardiovascular Diseases (CIBER-CV), Madrid, Spain
| | - O J Arias-Mutis
- Department of Physiology, School of Medicine, University of Valencia, Spain; Institute of Health Research INCLIVA, Valencia, Spain; Center for Biomedical Research Network on Cardiovascular Diseases (CIBER-CV), Madrid, Spain; Department of Biomedical Sciences, CEU Cardenal Herrera, Valencia, Spain
| | - M Aldasoro
- Department of Physiology, School of Medicine, University of Valencia, Spain; Institute of Health Research INCLIVA, Valencia, Spain
| | - J M Vila
- Department of Physiology, School of Medicine, University of Valencia, Spain; Institute of Health Research INCLIVA, Valencia, Spain
| | - E Serna
- Department of Physiology, School of Medicine, University of Valencia, Spain; Institute of Health Research INCLIVA, Valencia, Spain
| | - M D Mauricio
- Department of Physiology, School of Medicine, University of Valencia, Spain; Institute of Health Research INCLIVA, Valencia, Spain; Center for Biomedical Research Network on Cardiovascular Diseases (CIBER-CV), Madrid, Spain.
| |
Collapse
|
68
|
Saber FR, Salehi H, Khallaf MA, Rizwan K, Gouda M, Ahmed S, Zengin G, Zhang L, Rakmai J, Secomandi E, Lucini L, Simal-Gandara J. Limonoids: Advances in Extraction, Characterization, and Applications. FOOD REVIEWS INTERNATIONAL 2025:1-62. [DOI: 10.1080/87559129.2025.2456494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
|
69
|
Bie N, Li S, Liang Q, Zheng W, Xu S, Liu H, Zhang X, Wei Z, Yong T, Yang X, Gan L. Tumor-Repopulating Cell-Derived Microparticle-Based Therapeutics Amplify the Antitumor Effect through Synergistic Inhibition of Chemoresistance and Immune Evasion. Mol Pharm 2025; 22:733-746. [PMID: 39772575 DOI: 10.1021/acs.molpharmaceut.4c00709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Traditional chemotherapy often encounters failure attributed to drug resistance mediated by tumor-repopulating cells (TRCs) and chemotherapy-triggered immune suppression. The effective inhibition of TRCs and the mitigation of drug-induced immune suppression are pivotal for the successful chemotherapy. Here, TRC-derived microparticles (3D-MPs), characterized by excellent tumor-targeting and high TRC uptake properties, are utilized to deliver metformin and the chemotherapeutic drug doxorubicin ((DOX+Met)@3D-MPs). (DOX+Met)@3D-MPs efficiently enhance tumor accumulation and are highly internalized in tumor cells and TRCs. Additionally, (DOX+Met)@3D-MPs significantly decrease the chemotherapy-triggered upregulation in P-glycoprotein expression to enhance intracellular doxorubicin retention, resulting in increased chemotherapy sensitivity and immunogenic cell death in tumor cells and TRCs for improved antitumor immunity. Importantly, (DOX+Met)@3D-MPs also remarkably reduce chemotherapy-induced PD-L1 expression, efficiently alleviating immune suppression facilitated by the PD-L1/PD-1 axis to further enhance immunological response against malignancy. These results underscore the (DOX+Met)@3D-MPs' potential as a viable platform for augmenting the efficacy of antitumor therapies.
Collapse
Affiliation(s)
- Nana Bie
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qingle Liang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wenxia Zheng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyi Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Haojie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaojuan Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
70
|
Cheng D, Huang X, Shao B, Zhang C, Li X, Li M. Preventive efficacy of sprouting black soybean peptides on high-salt diet-induced hypertension in mice. J Food Sci 2025; 90:e70014. [PMID: 39961801 DOI: 10.1111/1750-3841.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/13/2024] [Accepted: 01/08/2025] [Indexed: 05/09/2025]
Abstract
This study examined possible mechanisms of action as well as the preventive and interventional effects of sprouting black soybean peptides (SBSPs) and black soybean peptides (BSPs) on hypertension in C57BL/6j mice that was brought on by a high-salt diet. BSP and SBSP were administered to mice in the black soybean peptide prevention (BSP-P) group and sprouting black soybean peptide prevention (SBSP-P) group starting 4 weeks prior to the high-salt diet, respectively. Mice in the black soybean peptide intervention (BSP-I) group and the sprouting black soybean peptide intervention (SBSP-I) group received oral doses of BSP and SBSP, respectively, together with a high-salt diet. The findings demonstrated that BSP-I, BSP-P, SBSP-I, and SBSP-P prevented the activation of localized angiotensin converting enzyme (ACE)/angiotensin II (Ang II) pathways in the kidneys and circulation, delayed the rise in blood pressure in mice, and preserved the functional nitric oxide/endothelin-1 (NO/ET-1) balance of endothelium. The inflammatory factors tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), and interleukin-6 (IL-6) had decreased plasma levels. Additionally, it improved fibrosis, renal edema phenomena, and cardiomegaly. Furthermore, the expression of genes related to the renin-angiotensin system and endothelial function is regulated by SBSP and BSP. On the other hand, compared to intervention effects, SBSP and BSP showed more noticeable preventive benefits on hypertension brought on by a high-salt diet. When all else was equal, SBSP was more effective than BSP at avoiding hypertension. As a result, this study offers theoretical backing for SBSP as a dietary supplement source for the prevention of high-salt diet-induced hypertension.
Collapse
Affiliation(s)
- Dawei Cheng
- Key Laboratory of Jianghuai Agricultural Product Fine Processing and Resource Utilization of Ministry of Agriculture and Rural Affairs, Anhui Engineering Research Center for High Value Utilization of Characteristic Agricultural Products, College of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Xinyu Huang
- Key Laboratory of Jianghuai Agricultural Product Fine Processing and Resource Utilization of Ministry of Agriculture and Rural Affairs, Anhui Engineering Research Center for High Value Utilization of Characteristic Agricultural Products, College of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Ben Shao
- Key Laboratory of Jianghuai Agricultural Product Fine Processing and Resource Utilization of Ministry of Agriculture and Rural Affairs, Anhui Engineering Research Center for High Value Utilization of Characteristic Agricultural Products, College of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Caihong Zhang
- Key Laboratory of Jianghuai Agricultural Product Fine Processing and Resource Utilization of Ministry of Agriculture and Rural Affairs, Anhui Engineering Research Center for High Value Utilization of Characteristic Agricultural Products, College of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Xueling Li
- Key Laboratory of Jianghuai Agricultural Product Fine Processing and Resource Utilization of Ministry of Agriculture and Rural Affairs, Anhui Engineering Research Center for High Value Utilization of Characteristic Agricultural Products, College of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Meiqing Li
- Key Laboratory of Jianghuai Agricultural Product Fine Processing and Resource Utilization of Ministry of Agriculture and Rural Affairs, Anhui Engineering Research Center for High Value Utilization of Characteristic Agricultural Products, College of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
71
|
El-Gammal GA, Ali GIE, Saif AS, Elbaz S, Fadl SE, Abu-Zahra NIS. The immunomodulatory and antioxidative effects of curcumin-supplemented diets against the isolated Aeromonas hydrophila in Oreochromisniloticus. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110077. [PMID: 39653178 DOI: 10.1016/j.fsi.2024.110077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
Worldwide, Aeromonas hydrophila (A. hydrophila) has become a significant foodborne pathogen, causing gastrointestinal and extra-intestinal infections in humans as well as severe economic losses in fish farms. Thus, we conducted this research in two experiments. The experiment 1 investigated the prevalence of A. hydrophila among Nile tilapia in different farms in Kafrelsheikh province and its pathogenicity by detecting some of its virulence genes before being used in the in vivo experiment. The experiment 2 was done to investigate the effect of dietary curcumin on growth performance, humoral immunity, antioxidants, and histopathology of Nile tilapia through a sixty-day feeding trial. Nile tilapia was divided into 3 groups, each in 3 replicates. Fish were given diets that included 0.0 (C, control), 1 % (10 g curcumin/kg diet; G1), and 2 % curcumin/kg diet (20 g curcumin/kg diet; G2). Moreover, the antibacterial effect of dietary curcumin against isolated bacteria in experiment 1 was investigated through experimental infection. The study's findings of experiment 1 indicated that the incidence of isolated A. hydrophila was 28 %. Moreover, the virulence aerolysin (aerA) and haemolysin (hlyA) genes were found in 100 % and 50 % of the isolates, respectively. The experiment 2 outcomes demonstrated that the growth-modulating impacts of dietary curcumin were notably noticed in final weight, feed intake, weight gain%, and specific growth rate without pathological lesions in the different organs. In addition, the white blood cell and lymphocyte counts significantly (P < 0.05) increased in Nile tilapia fed dietary curcumin. Similar improvements in humoral immunity (lysozyme and phagocytic activity) and antioxidants (catalase and superoxide dismutase) were seen in the 1 % curcumin group (G1). The experimental infection with a virulent strain of A. hydrophila that was previously isolated in experiment 1 showed a significant decrease in the morbidity and mortality rates in the 1 % curcumin treatment group (G1), followed by the 2 % group (G2). The present study concluded that under current experimental conditions, dietary curcumin at a dose of 10 g/kg diet proves effective, but the dose of 1 % may not be optimal for tilapia.
Collapse
Affiliation(s)
- Ghada A El-Gammal
- Bacteriology Unit, Kafrelsheikh Regional Lab, Animal Health Research Institute, Agricultural Research Center (ARC), Giza, Egypt.
| | - Gehan I E Ali
- Biochemistry, Nutritional Deficiency Diseases and Toxicology Unit, Kafrelsheikh Provincial Lab, Animal Health Research Institute, Agricultural Research Center (ARC), Giza, Egypt
| | - Aly S Saif
- Biochemistry, Nutritional Deficiency Diseases and Toxicology Unit, Kafrelsheikh Provincial Lab, Animal Health Research Institute, Agricultural Research Center (ARC), Giza, Egypt
| | - Shaimaa Elbaz
- Biochemistry, Nutritional Deficiency Diseases and Toxicology Unit, Kafrelsheikh Provincial Lab, Animal Health Research Institute, Agricultural Research Center (ARC), Giza, Egypt
| | - Sabreen E Fadl
- Biochemistry Department, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Nagwa I S Abu-Zahra
- Fish Diseases Unit, Kafrelsheikh Provincial Lab, Animal Health Research Institute (AHRI), Agriculture Research Center (ARC), Giza, Egypt
| |
Collapse
|
72
|
Xiong N, Liu S, Hu W, Liu Y, Ding X, Xu D, Ling Y, Fang M, Liu B, Zheng L, Zheng W, Wu B. Selenium yeast alleviates diquat-induced oxidative stress and testicular damage in roosters. Anim Reprod Sci 2025; 273:107760. [PMID: 39756356 DOI: 10.1016/j.anireprosci.2024.107760] [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: 10/07/2024] [Revised: 12/09/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025]
Abstract
Diquat (DQ) is a pro-oxidant that generates free radicals in cells through redox reactions, leading to the induction of oxidative stress. During the processes of growth and reproduction, poultry are particularly vulnerable to oxidative stress. Selenium yeast (SeY) serves as an organic selenium source characterized by high activity and low toxicity, imparting antioxidant effects. The objective of this study was to assess the protective effects of SeY against DQ-induced oxidative stress in rooster testicles.The results demonstrated that SeY pretreatment mitigated DQ-induced oxidative damage in the testes. This mitigation encompassed the alleviation of inhibited spermatogenesis, reduced spermatogenic cell abundance, and the alleviation of decreased expression of genes StAR, P450scc, and 3β-HSD which related to testosterone synthesis. Specifically, SeY pretreatment counteracted DQ-induced oxidative stress by activating the Nrf2/HO-1 antioxidant signaling pathway, enhancing the activity of antioxidant enzymes such as catalase (CAT) and total superoxide dismutase (T-SOD), and reducing the concentration of malondialdehyde (MDA). Furthermore, SeY pretreatment attenuated DQ-induced spermatogonia apoptosis by modulating the expression of apoptosis-related genes and proteins, including Bax, Bcl-2, Caspase3, and NF-κB. Additionally, SeY restored the proliferative capacity of spermatogenic cells by promoting the expression of the proliferation-related protein Ki67. The aforementioned findings signify that SeY effectively safeguards the testes against DQ-induced damage through mechanisms involving the reduction of oxidative stress, inhibition of apoptosis, promotion of proliferation, and enhancing the expression of testosterone synthesis related genes. This study lays a solid theoretical foundation for future research aimed at safeguarding the reproductive health of male poultry exposed to agricultural pesticides.
Collapse
Affiliation(s)
- Ningna Xiong
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China
| | - Sili Liu
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China
| | - Wei Hu
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China; Engineering Technology Research Center of Jiangxi Universities and Colleges for Selenium Agriculture, Yichun University, Yichun, Jiangxi 336000, China; Laboratory of Animal Pathogenic Microbiology, Yichun University, Yichun, Jiangxi 336000, China.
| | - Yamei Liu
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China
| | - Xiang Ding
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China
| | - Deming Xu
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China
| | - Yiqiang Ling
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China
| | - Manxin Fang
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China; Engineering Technology Research Center of Jiangxi Universities and Colleges for Selenium Agriculture, Yichun University, Yichun, Jiangxi 336000, China
| | - Ben Liu
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China; Engineering Technology Research Center of Jiangxi Universities and Colleges for Selenium Agriculture, Yichun University, Yichun, Jiangxi 336000, China; Laboratory of Animal Pathogenic Microbiology, Yichun University, Yichun, Jiangxi 336000, China
| | - Lucheng Zheng
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China; Engineering Technology Research Center of Jiangxi Universities and Colleges for Selenium Agriculture, Yichun University, Yichun, Jiangxi 336000, China
| | - Wenya Zheng
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China; Engineering Technology Research Center of Jiangxi Universities and Colleges for Selenium Agriculture, Yichun University, Yichun, Jiangxi 336000, China
| | - Bingyan Wu
- College of Life Science and Resources and Environment, Yichun University, Yichun, Jiangxi 336000, China; Engineering Technology Research Center of Jiangxi Universities and Colleges for Selenium Agriculture, Yichun University, Yichun, Jiangxi 336000, China
| |
Collapse
|
73
|
Wang X, Wang XQ, Luo K, Bai H, Qi JL, Zhang GX. Research Progress of Chinese Medicine Monomers in Treatment of Cholangiocarcinoma. Chin J Integr Med 2025; 31:170-182. [PMID: 39470920 DOI: 10.1007/s11655-024-4203-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 11/01/2024]
Abstract
Cholangiocarcinoma (CCA) is a malignant tumor originating from cholangiocytes. However, it remains unclear about the pathogenesis of this carcinoma, which may be related to multiple factors. Currently, CCA is mainly treated by surgery, chemotherapy, and radiotherapy. Among them, surgery is the only potentially curative option for CCA. Nevertheless, the high malignancy and asymptomatic nature of CCA may lead to poor treatment outcomes. It has been demonstrated that Chinese medicine (CM) plays a significant role in various antitumor applications. Meanwhile, CM exhibits fewer side effects and high availability. Moreover, the in vitro application of CM monomers has been explored in many domestic and foreign studies. This article mainly reviews the signaling pathways and molecular mechanisms of CM monomers in the treatment of CCA in recent years. These findings are expected to provide new insights into the treatment of CCA.
Collapse
Affiliation(s)
- Xiang Wang
- Department of General Surgery, the Second Hospital of Dalian Medical University, Dalian, Liaoning Province, 116027, China
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116021, China
- Hepatobiliary Surgery Department, Shandong Provincial Third Hospittal, Shandong University, Jinan, 250031, China
| | - Xiao-Qing Wang
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116021, China
| | - Kai Luo
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116021, China
| | - He Bai
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116021, China
| | - Jia-Lin Qi
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116021, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning Province, 116044, China
| | - Gui-Xin Zhang
- Department of General Surgery, the Second Hospital of Dalian Medical University, Dalian, Liaoning Province, 116027, China.
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning Province, 116044, China.
| |
Collapse
|
74
|
Shi X, Ma J, Liu W, Shen J, Xu G, Zhang J, Liu L. Pharmacokinetics Integrated With Network Pharmacology to Investigate the Potential Mechanism of Lu-Jiao Fang Inhibited Endothelial-to-Mesenchymal Transition in Pressure Overload-Induced Cardiac Fibrosis. Biomed Chromatogr 2025; 39:e6075. [PMID: 39775935 DOI: 10.1002/bmc.6075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
The aim of this study was to investigate the potential mechanism of Lu-Jiao Fang (LJF) inhibiting endothelial-to-mesenchymal transition (EndMT) in pressure overload-induced cardiac fibrosis. Pharmacokinetic behaviors of the ingredients of LJF were evaluated by LC-MS/MS analysis. Then putative pathways by which LJF regulates EndMT were analyzed by network pharmacology and verified in transverse aortic constriction-induced cardiac fibrosis rats. Loganin, morroniside, salidroside, isopsoralen, and psoralen showed higher plasma, left and right ventricular Cmax and AUC0-t values than hesperidin, specnuezhenide, and icariside II. Twenty-four potential targets related to EndMT were identified, which were mainly involved in relaxin signaling pathway. AKT1, TP53, MMP9, HIF1A, Snail1, and MMP2 were key therapeutic targets in protein-protein interaction network. LJF reversed cardiac dysfunction, left ventricular dilation, and fibrosis and significantly downregulated collagen type I and III and EndMT regulators (Snail1 and Twist1) mRNA expression. In relaxin signaling pathway, the RXFP1 protein expression increased by 22.52%, and the protein phosphorylation of Smad2 and Smad3 decreased by 33.52% and 12.79%, in response to the treatment with LJF. This study initially revealed the EndMT inhibition effects and molecular mechanisms of LJF in cardiac fibrosis, providing a reference basis for the promotion of LJF in the clinic.
Collapse
Affiliation(s)
- Xiaoli Shi
- Department of Pharmacy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingya Ma
- Department of Pharmacy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Liu
- Department of Pharmacy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Shen
- Department of Pharmacy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guanglin Xu
- Department of Pharmacy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianwei Zhang
- Department of Pharmacy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Liu
- Department of Pharmacy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
75
|
Joković N, Pešić S, Vitorović J, Bogdanović A, Sharifi-Rad J, Calina D. Glucosinolates and Their Hydrolytic Derivatives: Promising Phytochemicals With Anticancer Potential. Phytother Res 2025; 39:1035-1089. [PMID: 39726346 DOI: 10.1002/ptr.8419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/29/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
Recent research has increasingly focused on phytochemicals as promising anticancer agents, with glucosinolates (GSLs) and their hydrolytic derivatives playing a central role. These sulfur-containing compounds, found in plants of the Brassicales order, are converted by myrosinase enzymes into biologically active products, primarily isothiocyanates (ITCs) and indoles, which exhibit significant anticancer properties. Indole-3-carbinol, diindolylmethane, sulforaphane (SFN), phenethyl isothiocyanate (PEITC), benzyl isothiocyanate, and allyl isothiocyanate have shown potent anticancer effects in animal models, particularly in breast, prostate, lung, melanoma, bladder, hepatoma, and gastrointestinal cancers. Clinical studies further support the chemopreventive effects of SFN and PEITC, particularly in detoxifying carcinogens and altering biochemical markers in cancer patients. These compounds have demonstrated good bioavailability, low toxicity, and minimal adverse effects, supporting their potential therapeutic application. Their anticancer mechanisms include the modulation of reactive oxygen species, suppression of cancer-related signaling pathways, and direct interaction with tumor cell proteins. Additionally, semi-synthetic derivatives of GSLs have been developed to enhance anticancer efficacy. In conclusion, GSLs and their derivatives offer significant potential as both chemopreventive and therapeutic agents, warranting further clinical investigation to optimize their application in cancer treatment.
Collapse
Affiliation(s)
- Nataša Joković
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Strahinja Pešić
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Jelena Vitorović
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Andrija Bogdanović
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, Ecuador
- Department of Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
76
|
Cheng M, Ding F, Li L, Dai C, Sun X, Xu J, Chen F, Li M, Li X. Exploring the role of curcumin in mitigating oxidative stress to alleviate lipid metabolism disorders. Front Pharmacol 2025; 16:1517174. [PMID: 39950117 PMCID: PMC11822302 DOI: 10.3389/fphar.2025.1517174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Lipid metabolism plays a crucial role in maintaining homeostasis and overall health, as lipids are essential molecules involved in bioenergetic processes. An increasing body of research indicates that disorders of lipid metabolism can contribute to the development and progression of various diseases, including hyperlipidemia, obesity, non-alcoholic fatty liver disease (NAFLD), diabetes mellitus, atherosclerosis, and cancer, potentially leading to poor prognoses. The activation of the oxidative stress pathway disrupts lipid metabolism and induces cellular stress, significantly contributing to metabolic disorders. A well-documented crosstalk and interconnection between these metabolic disorders exists. Consequently, researchers have sought to identify antioxidant-rich substances in readily accessible everyday foods for potential use as complementary therapies. Curcumin, known for its anti-inflammatory and antioxidant properties, has been shown to enhance cellular antioxidant activity, mitigate oxidative stress, and alleviate lipid metabolism disorders by reducing reactive oxygen species (ROS) accumulation. These effects include decreasing fat deposition, increasing fatty acid uptake, and improving insulin sensitivity. A review of the existing literature reveals numerous studies emphasizing the role of curcumin in the prevention and management of metabolic diseases. Curcumin influences metabolic disorders through multiple mechanisms of action, with the oxidative stress pathway playing a central role in various lipid metabolism disorders. Thus, we aimed to elucidate the role of curcumin in various metabolic disorders through a unified mechanism of action, offering new insights into the prevention and treatment of metabolic diseases. Firstly, this article provides a brief overview of the basic pathophysiological processes of oxidative stress and lipid metabolism, as well as the role of oxidative stress in the pathogenesis of lipid metabolism disorders. Notably, the article reviews the role of curcumin in mitigating oxidative stress and in preventing and treating diseases associated with lipid metabolism disorders, including hyperlipidemia, non-alcoholic fatty liver disease (NAFLD), atherosclerosis, obesity, and diabetes, thereby highlighting the therapeutic potential of curcumin in lipid metabolism-related diseases.
Collapse
Affiliation(s)
- Maojun Cheng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Hosptial of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Fang Ding
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Hosptial of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Liyang Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Hosptial of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Changmao Dai
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Hosptial of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaolan Sun
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Hosptial of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jia Xu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Hosptial of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Feier Chen
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Hosptial of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Mingxiu Li
- Sichuan No. 2 Hosptial of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xueping Li
- Hosptial of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
77
|
Fu C, Yang X. Cardiac injury caused by iron overload in thalassemia. Front Pediatr 2025; 13:1514722. [PMID: 39931654 PMCID: PMC11808023 DOI: 10.3389/fped.2025.1514722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
Cardiac iron overload affects approximately 25% of patients with β-thalassemia major, which is associated with increased morbidity and mortality. Two mechanisms are responsible for iron overload in β-thalassemia: increased iron absorption due to ineffective erythropoiesis and blood transfusions. This review examines the mechanisms of myocardial injury caused by cardiac iron overload and role of various clinical examination techniques in assessing cardiac iron burden and functional impairment. Early identification and intervention for cardiac injury and iron overload in β-thalassemia have the potential to prevent and reverse or delay its progression in the early stages, playing a crucial role in its prognosis.
Collapse
Affiliation(s)
- Chunxi Fu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Xue Yang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
78
|
Dokduang H, Katenak A, Kittirat Y, Tomacha J, Promraksa B, Taing L, Na Nakorn S, Klanrit P, Namwat N, Titapun A, Loilome W. Chemical composition and anticancer potential of water extracts derived from ground powder of Thai germinated brown rice ( Oryza sativa L.). Nat Prod Res 2025:1-8. [PMID: 39862045 DOI: 10.1080/14786419.2025.2457017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 01/02/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Powdered germinated Thai rice (Oryza sativa L.) is widely utilised as a dietary supplement to support health and prevent diseases. This study investigated the bioactive compound profile of water extracts from beverage powder made from Thai germinated brown rice (GBRE) and assessed its anticancer effects on cholangiocarcinoma, lung cancer, and liver cancer cell lines. Proton nuclear magnetic resonance (1H-NMR) revealed 23 metabolites, including amino acids, sugar, phenolic compounds and nitrogenous compounds. Additionally, GBRE exhibited anticancer properties by effectively inhibiting cancer cell growth, inducing cell cycle arrest, and reducing cell migration. Our findings highlight the nutritional benefits and anticancer potential of germinated brown rice powder in impeding cancer cell progression. This study demonstrates the nutritional benefits and anticancer effects of germinated brown rice powder in inhibiting cancer cell progression. Incorporating germinated brown rice powder for a nutraceutical supplement can be served as a potential strategy for cancer prevention or therapeutic intervention.
Collapse
Affiliation(s)
- Hasaya Dokduang
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Faculty of Medicine, Mahasarakham University, Mahasarakham, Thailand
- Biomedical Science Research Unit, Mahasarakham University, Mahasarakham, Thailand
| | - Anukoon Katenak
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | | | - Jittima Tomacha
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Bundit Promraksa
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Leakkhaing Taing
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Kampong Speu Institute of Technology, Kampong Speu, Cambodia
| | - Saran Na Nakorn
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Poramate Klanrit
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of System Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nisana Namwat
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of System Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Attapol Titapun
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharin Loilome
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of System Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
79
|
Li Q, Yang Z, Lu H, Liu F, Zhou D, Zou Y. Astragalin Exerted Hypoglycemic Effect by Both Inhibiting α-Glucosidase and Modulating AMPK Signaling Pathway. Nutrients 2025; 17:406. [PMID: 39940264 PMCID: PMC11820219 DOI: 10.3390/nu17030406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND The hypoglycemic activity of mulberry leaf polyphenols has been widely studied, while its mechanism of action needs further elucidation. METHODS The inhibitory activity mechanism of astragalin on α-glucosidase was investigated with a combination of multispectroscopic techniques and molecular docking. The hypoglycemic pathway was further revealed with a high-glucose human hepatocellular carcinomas (HepG2) cell model. RESULTS The results indicated that astragalin inhibited α-glucosidase with IC50 of 154.5 µM, which was the highest in potency among the main polyphenols from mulberry leaves. Astragalin could bind to α-glucosidase with a single inhibition site and quench its endofluorescence with a static quenching mechanism. Astragalin changed the secondary structure of α-glucosidase, and the decreased α-helix content, representing the un-folding conformation, resulted in the decreased activity. The molecular docking further indicated that two sustainable hydrogen bonds were generated between astragalin and α-glucosidase residue Ser-88 and Tyr-133. The main driving forces to form the astragalin-α-glucosidase complex were the van der Waals force and hydrogen bond. Astragalin at a concentration of 80 µg/mL obtained the best hypoglycemic effect by activating the Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling pathway. CONCLUSIONS This study provides new insights into the potential utilization of astragalin-rich foods in the improvement of diabetes mellitus.
Collapse
Affiliation(s)
- Qian Li
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China; (Q.L.); (Z.Y.); (F.L.); (D.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China
| | - Zhangchang Yang
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China; (Q.L.); (Z.Y.); (F.L.); (D.Z.)
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Huijie Lu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China;
| | - Fan Liu
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China; (Q.L.); (Z.Y.); (F.L.); (D.Z.)
| | - Donglai Zhou
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China; (Q.L.); (Z.Y.); (F.L.); (D.Z.)
| | - Yuxiao Zou
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China; (Q.L.); (Z.Y.); (F.L.); (D.Z.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
80
|
Alharbi HOA, Alshebremi M, Babiker AY, Rahmani AH. The Role of Quercetin, a Flavonoid in the Management of Pathogenesis Through Regulation of Oxidative Stress, Inflammation, and Biological Activities. Biomolecules 2025; 15:151. [PMID: 39858545 PMCID: PMC11763763 DOI: 10.3390/biom15010151] [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: 12/12/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Quercetin, a flavonoid found in vegetables and fruits, has been extensively studied for its health benefits and disease management. Its role in the prevention of various pathogenesis has been well-documented, primarily through its ability to inhibit oxidative stress, inflammation, and enhance the endogenous antioxidant defense mechanisms. Electronic databases such as Google Scholar, Scopus, PubMed, Medline, and Web of Science were searched for information regarding quercetin and its role in various pathogeneses. The included literature comprised experimental studies, randomized controlled trials, and epidemiological studies related to quercetin, while editorials, case analyses, theses, and letters were excluded. It has been reported to have a wide range of health benefits including hepatoprotective, antidiabetic, anti-obesity, neuroprotective, cardioprotective, wound healing, antimicrobial, and immunomodulatory effects, achieved through the modulation of various biological activities. Additionally, numerous in vitro and in vivo studies have shown that quercetin's efficacies in cancer management involve inhibiting cell signaling pathways, such as inflammation, cell cycle, and angiogenesis, activating cell signaling pathways including tumor suppressor genes, and inducing apoptosis. This review aims to provide a comprehensive understanding of the health benefits of quercetin in various pathogeneses. Additionally, this review outlines the sources of quercetin, nanoformulations, and its applications in health management, along with key findings from important clinical trial studies. Limited clinical data regarding quercetin's safety and mechanism of action are available. It is important to conduct more clinical trials to gain a deeper understanding of the disease-preventive potential, mechanisms of action, safety, and optimal therapeutic dosages. Furthermore, more research based on nanoformulations should be performed to minimize/overcome the hindrance associated with bioavailability, rapid degradation, and toxicity.
Collapse
Affiliation(s)
| | | | | | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
81
|
Dai X, Liang B, Sun Y. Luteolin ameliorates rat model of metabolic syndrome-induced cardiac injury by apoptosis suppression and autophagy promotion via NR4A2/p53 regulation. BMC Complement Med Ther 2025; 25:14. [PMID: 39833877 PMCID: PMC11744851 DOI: 10.1186/s12906-025-04749-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Reduced cardiac autophagy, inflammation, and apoptosis contribute to cardiovascular complications caused by metabolic syndrome (MetS). It is documented that the nuclear receptor 4A2 (NR4A2) could modulate autophagy and apoptosis in cardiac complications. The aim of this investigation was to assess the therapeutic potential of luteolin, with documented beneficial properties, against MetS-associated cardiac injury. METHODS Forty male albino Wistar rats were divided into 5 groups randomly as controls, MetS, and MetS animals treated with luteolin (25, 50, 100 mg/kg ip). The animal's weight, blood pressure, lipid profile, tolerance to glucose and insulin, and cardiac histopathology were evaluated. Moreover, troponin T, creatine kinase-myocardial band (CK-MB), inflammatory profile (IL-6, IL-1β, TNF-α), transforming growth factor-β1 (TGF-β1), oxidative stress, and matrix metalloproteinase-9 (MMP-9) were analyzed to determine the cardiac state. Cardiac NR4A2 and p53, as well as apoptotic (B-cell leukemia/lymphoma 2 [BCL-2], Caspase [CASP]-3, and CASP-9) and autophagic mediators (Sequestosome-1/p62, Microtubule-associated protein 1 A/1B-light chain 3 [LC3], and Beclin-1) were measured by RT-qPCR and ELISA. RESULTS Luteolin remarkably restored MetS-induced biochemical derangements and related cardiac injury via the suppression of apoptosis, inflammation, and stress but promotion of autophagy (p-value < 0.001). CONCLUSION Current findings revealed the promising therapeutical properties of luteolin against MetS-associated cardiovascular risks.
Collapse
Affiliation(s)
- Xiyan Dai
- Department of Comprehensive, Maoming People's Hospital, Maoming, 525000, China
| | - Bo Liang
- Department of MRI, Maoming People's Hospital, Maoming, 525000, China
| | - Yaolin Sun
- Department of Cardiovascular Medicine, First Hospital of Northwest University, Xi'an, 710043, China.
| |
Collapse
|
82
|
Cammarota A, Balsano R, Pressiani T, Bozzarelli S, Rimassa L, Lleo A. The Immune-Genomics of Cholangiocarcinoma: A Biological Footprint to Develop Novel Immunotherapies. Cancers (Basel) 2025; 17:272. [PMID: 39858054 PMCID: PMC11763448 DOI: 10.3390/cancers17020272] [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: 11/29/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Cholangiocarcinoma (CCA) represents approximately 3% of all gastrointestinal cancers and is a highly heterogeneous and aggressive malignancy originating from the epithelial cells of the biliary tree. CCA is classified by anatomical location into intrahepatic (iCCA), extrahepatic (eCCA), gallbladder cancer (GBC), and ampullary cancers. Although considered a rare tumor, CCA incidence has risen globally, particularly due to the increased diagnosis of iCCA. Genomic and immune profiling studies have revealed significant heterogeneity within CCA, leading to the identification of molecular subtypes and actionable genetic alterations in 40-60% of cases, particularly in iCCA. Among these, FGFR2 rearrangements or fusions (7-15%) and IDH1 mutations (10-20%) are common in iCCA, while HER2 amplifications/overexpression are more frequent in eCCA and GBC. The tumor-immune microenvironment (TIME) of CCAs plays an active role in the pathogenesis and progression of the disease, creating a complex and plastic environment dominated by immune-suppressive populations. Among these, cancer-associated fibroblasts (CAFs) are a key component of the TIME and are associated with worse survival due to their role in maintaining a poorly immunogenic landscape through the deposition of stiff extracellular matrix and release of pro-tumor soluble factors. Improved understanding of CCA tumor biology has driven the development of novel treatments. Combination therapies of cisplatin and gemcitabine with immune checkpoint inhibitors (ICIs) have replaced the decade-long standard doublet chemotherapy, becoming the new standard of care in patients with advanced CCA. However, the survival improvements remain modest prompting research into more effective ways to target the TIME of CCAs. As key mechanisms of immune evasion in CCA are uncovered, novel immune molecules emerge as potential therapeutic targets. Current studies are exploring strategies targeting multiple immune checkpoints, angiogenesis, and tumor-specific antigens that contribute to immune escape. Additionally, the success of ICIs in advanced CCA has led to interest in their application in earlier stages of the disease, such as in adjuvant and neoadjuvant settings. This review offers a comprehensive overview of the immune biology of CCAs and examines how this knowledge has guided clinical drug development, with a focus on both approved and emergent treatment strategies.
Collapse
Affiliation(s)
- Antonella Cammarota
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, 20090 Pieve Emanuele, Italy; (R.B.); (L.R.)
| | - Rita Balsano
- Department of Biomedical Sciences, Humanitas University, 20090 Pieve Emanuele, Italy; (R.B.); (L.R.)
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (T.P.); (S.B.)
| | - Tiziana Pressiani
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (T.P.); (S.B.)
| | - Silvia Bozzarelli
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (T.P.); (S.B.)
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, 20090 Pieve Emanuele, Italy; (R.B.); (L.R.)
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (T.P.); (S.B.)
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, 20090 Pieve Emanuele, Italy; (R.B.); (L.R.)
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| |
Collapse
|
83
|
Lv M, Sun Q, Yu Y, Bao J. Nanocurcumin in myocardial infarction therapy: emerging trends and future directions. Front Bioeng Biotechnol 2025; 12:1511331. [PMID: 39845374 PMCID: PMC11750836 DOI: 10.3389/fbioe.2024.1511331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Myocardial infarction (MI) is the leading cause of morbidity and mortality worldwide. Curcumin has been observed to significantly reduce pathological processes associated with MI. Its clinical application is limited due to its low bioavailability, rapid degradation, and poor solubility. Advancements in nanotechnology can be used to enhance its therapeutic potentials in MI. Curcumin nano-formulation enhances its solubility, stability, and bioavailability, allowing more precise delivery to ischemic cardiac tissue. Curcumin nanoparticles have been observed to successfully reduce infarct size, maintain heart function by modulating essential molecular pathways in MI. Its liposomal formulations provide sustained release and higher tissue penetration with improved pharmacokinetics and enhanced therapeutic efficacy. Preclinical studies revealed that nanocurcumin drastically lower oxidative stress indicators, inflammatory cytokines, and cardiac damage. Micelles composed of polymers have demonstrated high biocompatibility and targeting capabilities with increased cardio-protective effects. Research and clinical trials are essential for comprehensive analysis and efficacy of curcumin-based nano-therapeutics in cardiovascular condition and lowering risk of MI.
Collapse
Affiliation(s)
- Mei Lv
- General Medicine Department, Yantaishan Hospital, Yantai, Shandong, China
| | - Qing Sun
- Department of Cardiology, Yantaishan Hospital, Yantai, Shandong, China
| | - Yilin Yu
- Preventive medicine, Shandong University, Jinan, Shandong, China
| | - Jinwei Bao
- Department of Cardiology, Yantaishan Hospital, Yantai, Shandong, China
| |
Collapse
|
84
|
Vageli DP, Doukas PG, Georgiou D, Prokopiou MP, Ladaki NE, Papadopoulou A, Doukas SG, Zacharouli K, Makaritsis KP, Ioannou M. HIF-1α and VEGF Immunophenotypes as Potential Biomarkers in the Prognosis and Evaluation of Treatment Efficacy of Atherosclerosis: A Systematic Review of the Literature. FRONT BIOSCI-LANDMRK 2025; 30:27004. [PMID: 39862086 DOI: 10.31083/fbl27004] [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: 10/16/2024] [Revised: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Hypoxia-inducible factor 1 alpha (HIF-1α) and its related vascular endothelial growth factor (VEGF) may play a significant role in atherosclerosis and their targeting is a strategic approach that may affect multiple pathways influencing disease progression. This study aimed to perform a systematic review to reveal current evidence on the role of HIF-1α and VEGF immunophenotypes with other prognostic markers as potential biomarkers of atherosclerosis prognosis and treatment efficacy. METHODS We performed a systematic review of the current literature to explore the role of HIF-1α and VEGF protein expression along with the relation to the prognosis and therapeutic strategies of atherosclerosis. We used the terms {"Atherosclerosis" [OR] "Atheroma" [OR] "atheromatous plaque" [OR] "plaque atherosclerotic"} [AND] {"HIF-1α"} [AND] {"VEGF"} from 2009 up to May 2024 and the Medline/Embase/PubMed database. We used methodological approaches to assess unbiased data [ROBIS (Risk of Bias in Systematic) tool]. We used study eligibility criteria, and data were collected and evaluated from original articles by two independent teams, judged by an independent reviewer, and reported by PRISMA (Preferred Reporting Items for Systematic reviews and Meta-Analyses) 2020. RESULTS We included 34 original studies investigating 650 human specimens, 21 different cell lines, and 9 animal models. Increased HIF-1α in vascular smooth muscle cells, macrophages, or endothelial cells, under hypoxia, chronic loss of nitric oxide (NO), or reduced micro ribonucleic acid (miRNA)-17 and miR-20, is associated with the upregulation of pro-inflammatory molecules, such as interleukin-1 beta (IL-1β) or tumor necrosis factor-alpha (TNF-α), increased migration inhibitory factor of macrophages, glycolytic flux, lipid accumulation, necroptosis via miR-383, and adverse effects in atherosclerosis and plaque vulnerability. However, increased HIF-1α in lymphocytes is associated with decreased interferon-gamma (IFN-γ) and a favorable prognosis. Increased VEGF in a coronary artery, activated macrophages, or chronic exposure to methamphetamine is associated with elevated levels of serum inflammatory cells (interleukin-18; IL18), p38 mitogen-activated protein kinase (MAPK) phosphorylation, lipopolysaccharide-induced tumor necrosis factor-alpha factor (LITAF), and signal transducer and activator of transcription 6 isoform B (STAT6B) overexpression, leading to atherosclerosis progression and plaque break. However, VEGF overexpression in serum is marginally associated with an elevated risk for atherosclerosis. In contrast, stable overexpression of VEGF in macrophages correlates with reduced hyperplasia after arterial injury, reduced foam cell formation, and attenuation of atherosclerosis progression. HIF-1α/VEGF immunophenotypes reflect atherosclerosis treatment efficacy using, among others, HIF-inhibitors, statins, polyphenols, miR-497-5p, methylation modification, adenosine receptor antagonists, natural products, or glycosides. CONCLUSION We present an overview of HIF-1α/VEGF expression in chronic inflammatory-related atherosclerosis disease. Exploring pathogenetic mechanisms and therapeutic options, we included several studies using variable methods to evaluate HIF-1α/VEGF immunophenotypes with controversial and innovative results. Data limitations may include the use of different survival methods. Our data support HIF-1α/VEGF immunophenotypes as potential biomarkers of atherosclerosis prognosis and treatment efficacy.
Collapse
Affiliation(s)
- Dimitra P Vageli
- Department of Neurology, Neuroscience and Regeneration Research Center Yale University School of Medicine & VA-CT, West Haven, CT 06516, USA
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Panagiotis G Doukas
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School/Saint Peter's University Hospital, New Brunswick, NJ 08901, USA
| | - Dimitrios Georgiou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Michailangelos P Prokopiou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Nefeli E Ladaki
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Androniki Papadopoulou
- Department of Anesthesiology, G. Gennimatas General Hospital, 54635 Thessaloniki, Greece
| | - Sotirios G Doukas
- Department of Medicine, Section of Gastroenterology and Hepatology, Rutgers-Robert Wood Johnson Medical School/Saint Peter's University Hospital, New Brunswick, NJ 08901, USA
| | - Konstantina Zacharouli
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Konstantinos P Makaritsis
- Department of Medicine & Research Laboratory of Internal Medicine, Faculty of Medicine, University of Thessaly/National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, 41110 Larissa, Greece
| | - Maria Ioannou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| |
Collapse
|
85
|
Luo X, Tai Q, Liu X, Zhou X, Li W, Liu H, Ding Z, Abudureyimu M. SQSTM1/p62 confers resistance of intrahepatic cholangiocarcinoma cells to 5-Fluorouracil by promoting Nrf2 nuclear translocation. Mol Cell Toxicol 2025. [DOI: 10.1007/s13273-024-00495-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 01/25/2025]
|
86
|
Huong PTM, Hanh TTH, Hong Quang T, Cuong NX, Lee DS, Nam NH, Minh CV. Diterpenoid and phenolic constituents from corn silk ( Zea mays) with PTP1B inhibitory activity. Nat Prod Res 2025; 39:323-330. [PMID: 37820035 DOI: 10.1080/14786419.2023.2265038] [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: 06/06/2023] [Accepted: 09/24/2023] [Indexed: 10/13/2023]
Abstract
Chemical investigation of corn silk resulted in the isolation of nine secondary metabolites, including a new ent-kaurane diterpenoid, zeamaysditerpene A (1) and eight known compounds, stigmaydene A (2), stigmaydene J (3), stigmaydene L (4), stigmane D (5), demethyltorosaflavone D (6), chrysoeriol 6-C-β-boivinopyranosyl-7-O-β-D-glucopyranoside (7), deoxypodophyllotoxin (8), and α-peltatin glucoside (9). Their structures were elucidated using a combination of spectroscopic methods, including 1D and 2D NMR and HRESIQTOF mass spectra. The absolute configuration of 1 was deduced by applying electronic circular dichroism (ECD) calculation method. Among the isolates, only 6 displayed significant inhibition against PTP1B activity in a dose-dependent manner, with an IC50 value of 10.7 ± 0.1 µM. Furthermore, molecular docking simulation was carried out to explore the action perspective of 6 inside the enzyme PTP1B. This finding suggests that 6 might be a potential lead for the development of a new anti-diabetic agent.
Collapse
Affiliation(s)
- Pham Thi Mai Huong
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Tran Thi Hong Hanh
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Tran Hong Quang
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Nguyen Xuan Cuong
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Dong-Sung Lee
- College of Pharmacy, Chosun University, Gwangju, Republic of Korea
| | - Nguyen Hoai Nam
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Chau Van Minh
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| |
Collapse
|
87
|
Ma Z, Sun J, Li Z, Huang S, Li B. AMDHD1 acts as a tumor suppressor and contributes to activation of TGF-β signaling pathway in cholangiocarcinoma. Cell Death Differ 2025; 32:162-176. [PMID: 39143229 PMCID: PMC11742690 DOI: 10.1038/s41418-024-01361-y] [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: 03/23/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a malignant tumor of the digestive system, characterized by its aggressive behavior and the absence of effective therapeutic biomarkers. Although recent studies have implicated AMDHD1 in tumor formation, its role in CCA development has been insufficiently explored. We utilized multiple bioinformatic datasets alongside 108 clinical samples to examine AMDHD1 expression in CCA. Then, in vitro and in vivo experiments were conducted to assess its impact on tumor growth and metastasis. Furthermore, proteomic analysis and immunoprecipitation mass spectrometry were employed to identify the downstream effectors of AMDHD1. We discovered that AMDHD1 was down-regulated in CCA and this down-regulation was associated with adverse clinicopathological features and prognosis. We also demonstrated that overexpression of AMDHD1 hindered G1/S progression in the cell cycle and promoted apoptosis, thereby inhibiting tumor growth and metastasis. Mechanistically, we found that AMDHD1 operated in a TGF-β-dependent manner and the inhibition of TGF-β signaling abrogated the effect of AMDHD1 overexpression on CCA cells. Specifically, AMDHD1 inhibited the ubiquitination and degradation of the SMAD4 protein through binding to the MH2 domain and synergistically enhanced SMAD2/3 phosphorylation, which activated of TGF-β signaling pathway and resulted in the suppression of CCA cell proliferation and migration. Our study identifies AMDHD1 as a significant prognostic biomarker and a tumor suppressor in CCA. It underscores the pivotal role of the AMDHD1/TGF-β signaling pathway in the development and progression of CCA.
Collapse
Affiliation(s)
- Zuyi Ma
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jia Sun
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhenchong Li
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Shanzhou Huang
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Southern Medical University, Guangzhou, China.
| | - Binglu Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
88
|
Zhang G, Liu X, Liang H, Wang X, Wang Q, Wang Q. Comprehensive Quality Evaluation of Traditional Chinese Medicine Guanxinjing Capsule Based on Fingerprint and Multicomponent Simultaneous Quantitative Method. J Sep Sci 2025; 48:e70059. [PMID: 39778053 DOI: 10.1002/jssc.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/30/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025]
Abstract
A novel dual-wavelength ultrahigh performance liquid chromatography (UHPLC) fingerprint was established, 56 common peaks were confirmed and attributed to the source of the medicinal materials, and 13 chromatographic peaks of them were identified by UHPLC quadrupole time-of-flight (Q-TOF)-MS/MS and UHPLC-UV method. Furthermore, a simple and sensitive HPLC-quadrupole trap (Q-TRAP)-MS/MS was developed for the simultaneous determination of 16 active components with electrospray ionization (ESI) source switching between positive and negative modes in a single run. The above two methods were successfully applied for the quality evaluation of Guanxinjing capsule (GXJC). Finally, statistical analysis including principal component analysis (PCA), hierarchical cluster analysis (HCA), and orthogonal partial least squares discriminate analysis (OPLS-DA) were applied, and 13 variables with great contribution to different groups of samples were screened. This study not only provides a rapid, accurate, and comprehensive qualitative and quantitative method for quality evaluation of GXJC but it also provides a reference workflow for the other traditional Chinese medicines.
Collapse
Affiliation(s)
- Guanhua Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, Shijiazhuang, China
- Department of Health Services, Logistics University of the Chinese People's Armed Police Force, Tianjin, China
| | - Xin Liu
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, Shijiazhuang, China
- Department of Pharmacy, Chengde Central Hospital, Chengde, Hebei, China
| | - Hao Liang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, Shijiazhuang, China
| | - Xu Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, Shijiazhuang, China
| | - Qi Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, Shijiazhuang, China
| | - Qiao Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
89
|
Rahmani S, Najdegerami E, Razi M, Nikoo M. Potential therapeutic effects of shrimp protein hydrolysates on NAFLD-induced infertility disorders: Insights into redox balance, heat shock protein expression, and chromatin compaction in male rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:158-169. [PMID: 39850116 PMCID: PMC11756727 DOI: 10.22038/ijbms.2024.76649.16589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/31/2024] [Indexed: 01/25/2025]
Abstract
Objectives Nonalcoholic fatty liver disease (NAFLD) is known to disrupt testicular anti-oxidant capacity, leading to oxidative stress (OS) that can negatively affect male fertility by damaging sperm DNA. Heat shock proteins (HSP70 and HSP90), in association with transitional proteins (TP1 and TP2), play crucial roles in protecting sperm DNA integrity in oxidative conditions. Whiteleg shrimp protein hydrolysates (HPs) exhibit anti-oxidant properties, prompting this study to explore the potential of HPs in ameliorating NAFLD-induced testicular damage. Materials and Methods The study divided rats into four groups: control, a group subjected to a high-fat diet (HFD) to induce NAFLD without supplementation, and two HFD-induced NAFLD groups receiving HP doses (20 and 300 mg/kg). After 70 days, the testicular total anti-oxidant capacity (TAC), malondialdehyde (MDA), glutathione (GSH), glutathione disulfide (GSSG), HSP70-2a, HSP90 expression, and TP mRNA levels were assessed. Results The results showed that HFD-induced NAFLD significantly increased GSH and MDA levels and disrupted the GSH/GSSG ratio (P<0.05) while also reducing HSP70-2a, HSP90, TP1, and TP2 expression (P<0.05). However, HP administration effectively restored testicular redox balance, reduced oxidative stress, and enhanced these protective proteins' expression compared to HFD (P<0.05). Conclusion NAFLD negatively affects the testicular redox system and HSP and TP expression, disrupting male fertility potential. In contrast, HP-treated rats showed a marked effect on NAFLD-induced damage by improving testicular anti-oxidant status and regulating the expression of HSPs and TP proteins. These findings suggest a potential therapeutic role for HP in safeguarding male fertility against the damaging effects of NAFLD.
Collapse
Affiliation(s)
- Somayyeh Rahmani
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | | | - Mazdak Razi
- Division of Comparative Histology and Embryology, Department of Basic Science, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Mehdi Nikoo
- Artemia & Aquaculture Research Institute, Urmia University, Urmia, Iran
| |
Collapse
|
90
|
Zhao ZX, Zou QY, Ma YH, Morris-Natschke SL, Li XY, Shi LC, Ma GX, Xu XD, Yang MH, Zhao ZJ, Li YX, Xue J, Chen CH, Wu HF. Recent progress on triterpenoid derivatives and their anticancer potential. PHYTOCHEMISTRY 2025; 229:114257. [PMID: 39209239 DOI: 10.1016/j.phytochem.2024.114257] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Cancer poses a significant global public health challenge, with commonly used adjuvant or neoadjuvant chemotherapy often leading to adverse side effects and drug resistance. Therefore, advancing cancer treatment necessitates the ongoing development of novel anticancer agents with diverse structures and mechanisms of action. Natural products remain crucial in the process of drug discovery, serving as a primary source for pharmaceutical leads and therapeutic advancements. Triterpenoids are particularly compelling due to their complex structures and wide array of biological activities. Recent research has demonstrated that naturally occurring triterpenes and their derivatives have the potential to serve as promising candidates for new drug development. This review aims to comprehensively explore the anticancer properties of triterpenoids and their synthetic analogs, with a focus on recent advancements. Various aspects, such as synthesis, phytochemistry, and molecular simulation for structure-activity relationship analyses, are summarized. It is anticipated that triterpenoid derivatives will emerge as notable anticancer agents following further investigation into their mechanisms of action and in vivo studies.
Collapse
Affiliation(s)
- Zi-Xuan Zhao
- Beijing Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Qiong-Yu Zou
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Key Laboratory of Hunan Higher Education for Western Hunan Medicinal Plant and Ethnobotany, Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Key Laboratory of Natural Products Research and Utilization in Wuling Mountain Area, Department of Chemistry & Chemical Engineering, Huaihua University, Huaihua, 418008, China
| | - Ying-Hong Ma
- Beijing Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Susan L Morris-Natschke
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Xiang-Yuan Li
- Beijing Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Lin-Chun Shi
- Beijing Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Guo-Xu Ma
- Beijing Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Xu-Dong Xu
- Beijing Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Mei-Hua Yang
- Beijing Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Zi-Jian Zhao
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Key Laboratory of Hunan Higher Education for Western Hunan Medicinal Plant and Ethnobotany, Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Key Laboratory of Natural Products Research and Utilization in Wuling Mountain Area, Department of Chemistry & Chemical Engineering, Huaihua University, Huaihua, 418008, China
| | - Yuan-Xiang Li
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Key Laboratory of Hunan Higher Education for Western Hunan Medicinal Plant and Ethnobotany, Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Key Laboratory of Natural Products Research and Utilization in Wuling Mountain Area, Department of Chemistry & Chemical Engineering, Huaihua University, Huaihua, 418008, China
| | - Jing Xue
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China.
| | - Chin-Ho Chen
- Antiviral Drug Discovery Laboratory, Surgical Oncology Research Facility, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Hai-Feng Wu
- Beijing Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China; Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Key Laboratory of Hunan Higher Education for Western Hunan Medicinal Plant and Ethnobotany, Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Key Laboratory of Natural Products Research and Utilization in Wuling Mountain Area, Department of Chemistry & Chemical Engineering, Huaihua University, Huaihua, 418008, China; Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
91
|
Bati K, Baeti PB, Gaobotse G, Kwape TE. Leaf extracts of
Euclea natalensis
A.D.C ameliorate biochemical abnormalities in high-fat-low streptozotocin-induced diabetic rats through modulation of the AMPK-GLUT4 pathway. EGYPTIAN JOURNAL OF BASIC AND APPLIED SCIENCES 2024; 11:232-252. [DOI: 10.1080/2314808x.2024.2326748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/29/2024] [Accepted: 02/28/2024] [Indexed: 01/03/2025]
Affiliation(s)
- Keagile Bati
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana
- Department of Biomedical Sciences, School of Medicine, University of Botswana, Gaborone, Botswana
| | - Phazha Bushe Baeti
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, School of Allied Health Professions, University of Botswana, Gaborone, Botswana
| | - Goabaone Gaobotse
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana
| | - Tebogo Elvis Kwape
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana
| |
Collapse
|
92
|
Awosika JA, Monge C, Greten TF. Integration of circulating tumor DNA in biliary tract cancer: the emerging landscape. Hepat Oncol 2024; 11:2403334. [PMID: 39881555 PMCID: PMC11486096 DOI: 10.1080/20450923.2024.2403334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/09/2024] [Indexed: 01/31/2025] Open
Abstract
Precision medicine has emerged as a cornerstone in cancer treatment revolutionizing our approach across malignancies. Molecular profiling of biliary tract cancers (BTCs) has changed the treatment landscape positively by prolonging survival in an aggressively fatal malignancy in its advanced stages. The acquisition of tissue tumor DNA for genomic analysis in BTC is often anatomically challenging, limited by quantity and quality. In response, ctDNA has emerged as a noninvasive means of molecular profiling. The utility of both plasma and bile ctDNA has been explored in several studies demonstrating the high mutation detection rates and the ability to isolate targetable mutations when present. In addition, the concordance between plasma and tissue DNA provides validity in utilizing ctDNA results to infer treatment decisions. Analysis of ctDNA in BTC has also provided prognostic information and facilitated evaluation of clonal evolution with ease of serial measurements. Insight into novel mechanisms of resistance to targeted therapies are being uncovered in ctDNA. As research endeavors continue to deepen our understanding in the field particularly in the space of ctDNA surveillance after curative intent, the tremendous progress made so far has enabled integration of ctDNA into the clinical practice of BTCs.
Collapse
Affiliation(s)
- Joy A Awosika
- Gastrointestinal Malignancies Section, Thoracic & GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD20892, USA
| | - Cecilia Monge
- Gastrointestinal Malignancies Section, Thoracic & GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD20892, USA
| | - Tim F Greten
- Gastrointestinal Malignancies Section, Thoracic & GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD20892, USA
| |
Collapse
|
93
|
Aghdam MA, Pagán A, García-Estañ J, Atucha NM. Evaluation of the Effects of Mulberry Leaf Extracts Morus alba L. on Cardiovascular, Renal, and Platelet Function in Experimental Arterial Hypertension. Nutrients 2024; 17:49. [PMID: 39796483 PMCID: PMC11722676 DOI: 10.3390/nu17010049] [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: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
INTRODUCTION Numerous epidemiological studies have demonstrated that consuming foods rich in polyphenols and flavonoids can have beneficial effects on various diseases, including arterial hypertension (HTN). Recent research from our laboratory has shown that certain flavonoids exhibit antihypertensive properties in several animal models of HTN. Our objective was to evaluate the effect of Morus alba L. (white mulberry) extracts in an experimental HTN model characterized by nitric oxide (NO) deficiency. METHODS Male Sprague-Dawley rats were divided into four groups: a control group, hypertensive rats treated with an NO synthesis inhibitor (L-NAME) in drinking water for six weeks, L-NAME rats treated with Morus alba L. extract, and L-NAME rats treated simultaneously with captopril. After six weeks of treatment, we measured blood pressure, endothelial vascular function in the aorta, and platelet aggregation function. RESULTS Morus alba L. extract partially prevented the development of arterial hypertension due to NO deficiency, although it did not completely normalize blood pressure as captopril did. The extract reduced the excessive vasoconstrictor response to phenylephrine in aortic rings and improved vasodilation in response to acetylcholine, with both effects dependent on increased NO production. Morus alba L. extract also reduced the increased platelet aggregation in response to ADP and collagen in hypertensive animals, although it did not fully normalize this function. CONCLUSIONS Morus alba L. extract demonstrates antihypertensive effects, improves vascular reactivity, and reduces platelet aggregation in a model of arterial hypertension. These effects are primarily related to an increase in nitric oxide activity.
Collapse
Affiliation(s)
| | | | - Joaquín García-Estañ
- Departamento Fisiología, Facultad Medicina, Instituto Murciano de Investigación Biosanitaria, Universidad de Murcia, 30120 Murcia, Spain (A.P.); (N.M.A.)
| | | |
Collapse
|
94
|
Eryilmaz IE, Colakoglu Bergel C, Arioz B, Huriyet N, Cecener G, Egeli U. Luteolin induces oxidative stress and apoptosis via dysregulating the cytoprotective Nrf2-Keap1-Cul3 redox signaling in metastatic castration-resistant prostate cancer cells. Mol Biol Rep 2024; 52:65. [PMID: 39699825 DOI: 10.1007/s11033-024-10178-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND The treatment of metastatic castration-resistant prostate cancer (mCRPC) is still challenging clinically. Due to the refractor and highly metastatic phenotype of mCRPC, novel therapy strategies need to be investigated. Luteolin, a promising anticancer agent with various biological targets in many cancer types, also has a pro-oxidant effect that selectively triggers ROS and apoptosis. In recent years, among its ROS-mediated mechanisms, the inhibitory effect of luteolin on the nuclear factor-E2-related factor 2 (Nrf2), the main ROS scavenger protein in cancer cells, has been reported. However, no evidence exists that luteolin potentially regulates the Nrf2 or its regulator signaling pathway, Nrf2-Keap1-Cul3 axis, concerning its pro-oxidant effects associated with ROS-triggered apoptosis in any PCa cells or tumor model. METHODS AND RESULTS In the present study, we investigated for the first time whether the anticancer effect of luteolin is associated with pro-oxidant activity via the regulation of the Nrf2-Keap1-Cul3 redox signaling in PC3 and DU145 mCRPC cells. The results showed that luteolin significantly caused more cytotoxic, apoptotic, and pro-oxidant effects in a dose-dependent manner in mCRPC cells than in WPMY-1 normal prostate fibroblast cells for 72 h. Moreover, significant inhibition of Nrf2-Keap1-Cul3 redox signaling has occurred in response to increasing doses of luteolin in mCRPC cells. CONCLUSIONS The current study put forth the potential pro-oxidant inhibitory effect of luteolin on the Nrf2-Keap1-Cul3 axis in mCRPC cells for the first time. Thus, luteolin might be an attractive therapy strategy with an inhibitory effect on the cytoprotective Nrf2-Keap1-Cul3 redox signaling for treating mCRPC.
Collapse
Affiliation(s)
- Isil Ezgi Eryilmaz
- Medical Biology Department, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.
| | | | - Bilge Arioz
- Medical Biology Department, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Nuseybe Huriyet
- Medical Biology Department, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Gulsah Cecener
- Medical Biology Department, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Unal Egeli
- Medical Biology Department, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
95
|
Boonla O, Booranasuksakul U, Padkao T, Phoemsapthawee J, Tangwattanachuleeporn M, Koowattanatianchai S, Prasertsri P. Effects of 4-week Eri silkworm cornflakes supplementation on oxidative stress and antioxidant status in male university athletes: A preliminary crossover study. Nutr Health 2024:2601060241302387. [PMID: 39699021 DOI: 10.1177/02601060241302387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Aim: This study among moderately fit male athletes investigated changes in oxidative stress and antioxidant levels after supplementation with Eri silkworm cornflakes. Method: Male university athletes (n = 18, age 19-21 years) were enrolled in a randomized crossover trial. They participated in either the supplementation period (consuming 0.20 g protein/kg body weight Eri silkworm daily) or the control period (no supplementation). Each period lasted 4 weeks, with a washout period of 4 weeks in between. Results: Compared to the control period, reduced glutathione concentration (p = 0.019) and reduced/oxidized glutathione ratio (p < 0.001) were significantly higher, and oxidized glutathione (p = 0.018) and malondialdehyde (p = 0.002) concentrations were significantly lower in the supplementation period. Conclusion: Eri silkworm supplementation as cornflakes for 4 weeks improved oxidative stress and antioxidant status in male university athletes. This supplement may be an alternative to support optimal antioxidant status for athletic individuals. Thai Clinical Trials Registry (TCTR): TCTR20180303003.
Collapse
Affiliation(s)
- Orachorn Boonla
- Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | | | - Tadsawiya Padkao
- Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Jatuporn Phoemsapthawee
- Department of Sports Science, Faculty of Sports and Health Science, Kasetsart University, Nakhon Pathom, Thailand
| | | | - Sukrisd Koowattanatianchai
- Division of Cardiology, Department of Medicine, Burapha Hospital, Burapha University, Chonburi, Thailand
| | | |
Collapse
|
96
|
Edirisinghe O, Ternier G, Alraawi Z, Suresh Kumar TK. Decoding FGF/FGFR Signaling: Insights into Biological Functions and Disease Relevance. Biomolecules 2024; 14:1622. [PMID: 39766329 PMCID: PMC11726770 DOI: 10.3390/biom14121622] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Fibroblast Growth Factors (FGFs) and their cognate receptors, FGFRs, play pivotal roles in a plethora of biological processes, including cell proliferation, differentiation, tissue repair, and metabolic homeostasis. This review provides a comprehensive overview of FGF-FGFR signaling pathways while highlighting their complex regulatory mechanisms and interconnections with other signaling networks. Further, we briefly discuss the FGFs involvement in developmental, metabolic, and housekeeping functions. By complementing current knowledge and emerging research, this review aims to enhance the understanding of FGF-FGFR-mediated signaling and its implications for health and disease, which will be crucial for therapeutic development against FGF-related pathological conditions.
Collapse
Affiliation(s)
- Oshadi Edirisinghe
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Gaëtane Ternier
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| | - Zeina Alraawi
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| | - Thallapuranam Krishnaswamy Suresh Kumar
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA;
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| |
Collapse
|
97
|
Bunbamrung N, Intaraudom C, Dramae A, Boonyuen N, Choowong W, Rachtawee P, Thawai C, Pittayakhajonwut P. Antimalarial and antimicrobial substances from the endophytic fungus Chaetomium globosum BCC71876. Nat Prod Res 2024:1-10. [PMID: 39676752 DOI: 10.1080/14786419.2024.2440800] [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: 08/25/2024] [Revised: 11/13/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
Three previously undescribed compounds including 2',3'-dihydroxy-4,5',4″-trimethoxy-p-terphenyl (1), 5-methoxyhydroperoxyco-chliodinol (2), and 5-(3',3'-dimethylallyl)indole-3-oxoacetic acid (17), along with fifteen known compounds (3 - 16 and 18), were isolated from the endophytic fungus, Chaetomium globosum BCC71876. The chemical structures were determined by NMR spectral information as well as the comparison with those closely related compounds reported in the literature. The compounds isolated were evaluated for biological properties including antimalarial, anti-TB, anti-plant pathogenic fungal, antibacterial, and cytotoxicity. The isolated compounds exhibited a wide range of biological activities against the tested cell lines and showed very low cytotoxicity against both cancerous (MCF-7 and NCI-H187) and non-cancerous (Vero) cells, except for compounds 5, 10, 11, 17, and 18 (IC50 0.47 - 114.8 µM). Cochliodinol (5) had the strongest antimalarial activity (IC50 4.39 µM), whereas chaetomugilin J (11) showed the strongest anti-TB activity (MIC 3.13 µg/mL).
Collapse
Affiliation(s)
- Nantiya Bunbamrung
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Chakapong Intaraudom
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Aibrohim Dramae
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Nattawut Boonyuen
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Wilunda Choowong
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Pranee Rachtawee
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Chitti Thawai
- Department of Biology, Faculty of Science, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
- Actinobacterial Research Unit, Faculty of Science, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Pattama Pittayakhajonwut
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| |
Collapse
|
98
|
Sobańska AW, Sobański AM. Organic Sunscreens-Is Their Placenta Permeability the Only Issue Associated with Exposure During Pregnancy? In Silico Studies of Sunscreens' Placenta Permeability and Interactions with Selected Placental Enzymes. Molecules 2024; 29:5836. [PMID: 39769924 PMCID: PMC11728689 DOI: 10.3390/molecules29245836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
One of the functions of placenta is to protect the fetus against harmful xenobiotics. Protective mechanisms of placenta are based on enzymes, e.g., antioxidant enzymes from the glutathione S-transferases group (GST) or human N-acetyltransferase 2 (NAT2). Many organic sunscreens are known to cross biological barriers-they are detected in mother's milk, semen, umbilical cord blood or placental tissues. Some organic sunscreens are able to cross the placenta and to interfere with fetal development; they are known or suspected endocrine disruptors or neurotoxins. In this study, 16 organic sunscreens were investigated in the context of their placenta permeability and interactions with gluthatione S-transferase and human N-acetyltransferase 2 enzymes present in the human placenta. Binary permeability models based on discriminant analysis and artificial neural networks proved that the majority of studied compounds are likely to cross the placenta by passive diffusion. Molecular docking analysis suggested that some sunscreens show stronger affinity for glutathione S-transferase and human N-acetyltransferase 2 that native ligands (glutathione and Coenzyme A for GST and NAT2, respectively)-it is therefore possible that they are able to reduce the enzyme's protective activity. It was established that sunscreens bind to the studied enzymes mainly by alkyl, hydrogen bonds, van der Waals, π-π, π-alkyl and π-sulfur interactions. To conclude, sunscreens may become stressors affecting humans by different mechanisms and at different stages of development.
Collapse
Affiliation(s)
- Anna W. Sobańska
- Department of Analytical Chemistry, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | | |
Collapse
|
99
|
Lv B, Xing S, Wang Z, Zhang A, Wang Q, Bian Y, Pei Y, Sun H, Chen Y. NRF2 inhibitors: Recent progress, future design and therapeutic potential. Eur J Med Chem 2024; 279:116822. [PMID: 39241669 DOI: 10.1016/j.ejmech.2024.116822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a crucial transcription factor involved in oxidative stress response, which controls the expression of various cytoprotective genes. Recent research has indicated that constitutively activated NRF2 can enhance patients' resistance to chemotherapy drugs, resulting in unfavorable prognosis. Therefore, the development of NRF2 inhibitors has emerged as a promising approach for overcoming drug resistance in cancer treatment. However, there are limited reports and reviews focusing on NRF2 inhibitors. This review aims to provide a comprehensive analysis of the structure and regulation of the NRF2 signaling pathway, followed by a comprehensive review of reported NRF2 inhibitors. Moreover, the current design strategies and future prospects of NRF2 inhibitors will be discussed, aiming to establish a foundation for the development of more effective NRF2 inhibitors.
Collapse
Affiliation(s)
- Bingbing Lv
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Zhiqiang Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Ao Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Qinjie Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yaoyao Bian
- Jiangsu Provincial Engineering Center of TCM External Medication Researching and Industrializing, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yuqiong Pei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
100
|
Chen F, Xue Y, Zhang W, Zhou H, Zhou Z, Chen T, YinWang E, Li H, Ye Z, Gao J, Wang S. The role of mitochondria in tumor metastasis and advances in mitochondria-targeted cancer therapy. Cancer Metastasis Rev 2024; 43:1419-1443. [PMID: 39307891 PMCID: PMC11554835 DOI: 10.1007/s10555-024-10211-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/08/2024] [Indexed: 11/05/2024]
Abstract
Mitochondria are central actors in diverse physiological phenomena ranging from energy metabolism to stress signaling and immune modulation. Accumulating scientific evidence points to the critical involvement of specific mitochondrial-associated events, including mitochondrial quality control, intercellular mitochondrial transfer, and mitochondrial genetics, in potentiating the metastatic cascade of neoplastic cells. Furthermore, numerous recent studies have consistently emphasized the highly significant role mitochondria play in coordinating the regulation of tumor-infiltrating immune cells and immunotherapeutic interventions. This review provides a comprehensive and rigorous scholarly investigation of this subject matter, exploring the intricate mechanisms by which mitochondria contribute to tumor metastasis and examining the progress of mitochondria-targeted cancer therapies.
Collapse
Affiliation(s)
- Fanglu Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yucheng Xue
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wenkan Zhang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hao Zhou
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhiyi Zhou
- The First People's Hospital of Yuhang District, Hangzhou, Zhejiang, China
| | - Tao Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Eloy YinWang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hengyuan Li
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Shengdong Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|