1
|
Ogunro OB. An updated and comprehensive review of the health benefits and pharmacological activities of hesperidin. Biochem Biophys Res Commun 2025; 772:151974. [PMID: 40414011 DOI: 10.1016/j.bbrc.2025.151974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVES This review aims to comprehensively assess the health benefits and pharmacological activities of hesperidin, a flavonoid commonly found in citrus fruits. It consolidates recent research findings to provide insights into hesperidin's diverse health-promoting effects. KEY FINDINGS Hesperidin has gained significant attention recently for its notable pharmacological activities and potential health benefits. Studies reveal its antioxidant properties, protecting cells from oxidative damage, and its anti-inflammatory effects, inhibiting pro-inflammatory cytokines and enzymes. Also, hesperidin shows promise in cardiovascular health by reducing blood pressure and cholesterol levels and enhancing endothelial function. It also exhibits anticancer potential by hindering cell proliferation, inducing apoptosis, and suppressing tumour growth. Moreover, hesperidin demonstrates neuroprotective effects, potentially mitigating neuroinflammation and oxidative stress associated with neurodegenerative diseases. Furthermore, it displays beneficial effects in metabolic disorders such as diabetes, obesity, and fatty liver disease by influencing glucose metabolism, lipid profile, and insulin sensitivity. SUMMARY Hesperidin exhibits a wide range of health benefits and pharmacological activities, making it a promising candidate for therapeutic interventions in various diseases. Its antioxidant, anti-inflammatory, cardiovascular, anticancer, neuroprotective, and metabolic effects underscore its potential as a valuable natural compound for promoting health and preventing chronic diseases.
Collapse
Affiliation(s)
- Olalekan Bukunmi Ogunro
- Drug Discovery, Toxicology, and Pharmacology Research Laboratory, Department of Biological Sciences, KolaDaisi University, Ibadan, Nigeria.
| |
Collapse
|
2
|
Eissa LA, Marawan AM, Marawan ME, Abass SA. Autophagy in disease management: Exploring the potential of natural products as targeted therapies. Pathol Res Pract 2025; 272:156077. [PMID: 40516139 DOI: 10.1016/j.prp.2025.156077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 06/01/2025] [Accepted: 06/08/2025] [Indexed: 06/16/2025]
Abstract
Autophagy is a vital cellular process that degrades and recycles intracellular components via lysosomes, playing a key role in maintaining cellular homeostasis. Alteration of this mechanism has been implicated in the occurrence and progression of numerous diseases, including cancer, neurodegenerative disorders, cardiovascular conditions, and microbial and viral infections. Recent studies have identified several mutations affecting autophagy-related genes and elucidated how defective degradation of specific substrates contributes to disease mechanisms. Natural products are gaining attention for their ability to modulate autophagy through several molecular targets. Herin, we highlight the complicated role of autophagy in disease pathogenesis. We also illustrate how natural products may offer therapeutic value by targeting autophagy in different pathological contexts.
Collapse
Affiliation(s)
- Laila A Eissa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Amani M Marawan
- Faculty of Veterinary Medicine, Delta University for Science and Technology, Gamsa 11152, Egypt; Shoha Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35738, Egypt.
| | - Mohamed E Marawan
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt.
| | - Shimaa A Abass
- Department of Biochemistry, Faculty of Pharmacy, Kaferelsheikh University, Kaferelsheikh 33516, Egypt; Biochemistry Department, Faculty of Pharmacy, Menoufia National University, Menoufia, Egypt.
| |
Collapse
|
3
|
Abdel-Maksoud MS, Alatawi RA, Albalawi SSA, Alrashidi MN, Abo-Dya NE, Elsherbiny N, Ragab YM, Awaji AA, El-Sherbiny M, Elfadil H, Abd-Alhaseeb MM. Diacerein's antiproliferative effects alone and with 5-fluorouracil in an Ehrlich solid tumour model: Molecular docking, molecular dynamics Simulation studies, and experimental Verification. Eur J Pharmacol 2025; 996:177564. [PMID: 40157706 DOI: 10.1016/j.ejphar.2025.177564] [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: 12/15/2024] [Revised: 03/16/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
The current study used an experimental model of mammary gland carcinoma to assess the chemo-sensitizing effectiveness of the combined administration of diacerein and 5-Fluorouracil (5-FU). With docking scores of -8.1, -7.6, and -9.2 kcal/mol, respectively, the molecular docking experiments showed that diacerein exhibits significant binding affinities to Caspase-3, NF-κB, and AKT1. Molecular dynamics Simulations revealed that diacerein has favourable binding free energy (ΔGbind) of -26.7 kcal/mol for Caspase-3, -24.2 kcal/mol for NF-κB, and -39.9 kcal/mol for AKT1, combined with low root mean square deviation (RMSD) values of 3.1 Å, 1.6 Å, and 2.1 Å for the three targets respectively. To validate these findings in vivo, Ehrlich solid tumor (EST) was induced in female Swiss mice. Four groups of animals were randomly assigned: EST + vehicle, EST + 5-FU, EST + diacerein, and EST + combination. Diacerein and 5-FU combination treatment increased EST mice's life span and reduced the solid tumor's weight and volume. Furthermore, diacerein and 5-FU combination significantly suppressed oxidative stress, inhibited AKT phosphorylation, decreased downstream inflammation (NF-κB, TNF-α, IL-1β), and increased apoptosis by modulating Bax, Bcl2, P53, and caspase-3 levels in tumor tissues. In conclusion, by inhibiting the AKT/NF-κB axis, diacerein and 5-FU combination showed possible antiproliferative effectiveness in the EST model.
Collapse
Affiliation(s)
- Mohamed S Abdel-Maksoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.
| | | | | | | | - Nader E Abo-Dya
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia.
| | - Nehal Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia.
| | - Yasser M Ragab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, North Sinai, Egypt; Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Aeshah A Awaji
- Department of Biology, Faculty of Science, University College of Taymaa, University of Tabuk, 71491, Saudi Arabia.
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia.
| | - Hassabelrasoul Elfadil
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.
| | - Mohammad M Abd-Alhaseeb
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt; Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
4
|
Al-Tantawy SM, Eraky SM, Eissa LA. Novel therapeutic target for diabetic kidney disease through downregulation of miRNA-192-5p and miRNA-21-5p by celastrol: implication of autophagy, oxidative stress, and fibrosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6915-6928. [PMID: 39702603 DOI: 10.1007/s00210-024-03669-5] [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: 10/17/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024]
Abstract
One of the most common microvascular effects of diabetes mellitus (DM) that may result in end-stage renal failure is diabetic kidney disease (DKD). Current treatments carry a substantial residual risk of disease progression regardless of treatment. By modulating various molecular targets, pentacyclic triterpenoid celastrol has been found to possess curative properties in the treatment of diabetes and other inflammatory diseases. Therefore, the present study investigated whether celastrol has anti-inflammatory, antioxidant, and antifibrotic effects as a natural compound against experimental DKD. Streptozotocin (55 mg/kg) was utilized for inducing DKD in a rat model. Antioxidant enzymes and renal function tests were assessed in serum samples. In kidney homogenate, relative miRNA-192-5p and miRNA-21-5p gene expressions were measured. Furthermore, using real-time PCR to evaluate the gene expressions of nucleus erythroid 2-related factor-2 (Nrf-2), matrix metalloproteinase-2 (MMP-2), proapoptotic caspase-3, antiapoptotic Bcl-2, LC-3, and Beclin-1. Moreover, the transforming growth factor β1 (TGF-β1), LC-3, Bcl-2, caspase-3 and NADPH oxidase 4 (NOX4) renal expressions were assessed semi-quantitatively using immunohistochemistry. Seven weeks of celastrol (1.5 mg/kg/day) treatment significantly ameliorated DKD. Celastrol improves kidney functions. Moreover, celastrol treatment demonstrated potent antioxidant effect. The mechanism of apoptosis resulting from the administration of celastrol included the modulation of Bcl-2 and caspase-3 expression in the kidney. Celasterol administration leads to an increase in LC-3 and Beclin-1 renal expression that resulting in autophagy. Celastrol treatment improved renal fibrosis by decreasing TGF-β1 and MMP-2 renal expression. These antifibrotic effects could be due to their ability to inhibit miRNA-192-5p and miRNA-21-5p expression in renal tissues. Celastrol exerts a renoprotective effect by targeting miRNA-21 and miRNA-192, as well as their downstream pathways, such as autophagy, apoptosis, and fibrosis.
Collapse
Affiliation(s)
- Samar M Al-Tantawy
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Salma M Eraky
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Laila A Eissa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
5
|
Tufail P, Anjum S, Siddiqui BS, Pizzi M, Jahan H, Choudhary MI. Nitrovanillin derivative ameliorates AGE-RAGE nexus associated inflammation: A step towards the amelioration of vascular complications under diabetic environment. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167784. [PMID: 40058471 DOI: 10.1016/j.bbadis.2025.167784] [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: 10/28/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
INTRODUCTION Advanced glycation endproducts (AGEs) are implicated in various pathological conditions, including diabetes, inflammation, and cardiovascular diseases. Methylglyoxal (MGO), a potent glycation agent, leads to the formation of MGO-derived AGEs, which promote structural and functional anomalies in various cellular and tissues proteins. AGEs stimulate the proliferation of monocytes, and induce a pro-inflammatory state through AGE-RAGE interactions, triggering oxidative stress, and inflammatory condition that contribute to the progression of atherosclerosis, and other diabetic complications. OBJECTIVE The current study was aimed to explore the antioxidant and anti-inflammatory properties of a series of novel antiglycation compounds, nitrovanillin derivatives, by modulating the AGEs-stimulated intracellular signaling pathways involved in inflammation. METHODS The preliminary safety profile of nitrovanillin derivatives was assessed by using human hepatocytes (HepG2), and monocytes (THP-1) cell lines via MTT, and WST-1 assays, respectively. Antioxidant activity of the compounds was determined by using DCFH-DA technique. Western blotting, immunocytochemistry, and ELISA methods were employed to assess the levels of pro-inflammatory markers (RAGE, COX-1, COX-2, NF-κB, and PGE2) in MGO-AGEs stimulated THP-1 monocytes. RESULT Among the nitrovanillin derivatives 1-11, only compound 2, ((E)-2-methoxy-6-nitro-4-(((2-(trifluoromethyl)phenyl)imino)methyl)phenol), was found non-toxic to HepG2, and THP-1 cells. Compound 2 lowered the MGO-AGEs-induced reactive oxygen species (ROS) production by inhibiting the upstream signaling of NADPH oxidase and MAPK-p38, which subsequently inhibited the NF-κB activation in THP-1 monocytes. Compound 2 also reversed the AGEs-mediated COX-1 suppression, COX-2 upregulation, and PGE2 production by blocking the AGE-RAGE ligation in THP-1 monocytes. CONCLUSION In conclusion, nitrovanillin 2 ((E)-2-methoxy-6-nitro-4-(((2-(trifluoromethyl)phenyl)imino)methyl)phenol) is a potential candidate for mitigating MGO-AGEs mediated vasculopathy by the inhibition of AGE-RAGE-p38/NF-κB nexus in THP-1 monocytes. It may offer a therapeutic option for the patients with diabetes and chronic inflammatory vascular complications, and thus offering new avenues for treatment development.
Collapse
Affiliation(s)
- Priya Tufail
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Sajjad Anjum
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Bina Shaheen Siddiqui
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Humera Jahan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Komplek Kampus C, JI. Mulyorejo, Surabaya 60115, Indonesia.
| | - M Iqbal Choudhary
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Komplek Kampus C, JI. Mulyorejo, Surabaya 60115, Indonesia.
| |
Collapse
|
6
|
Hsiao C, Liao WC, Li JP, Chou YC, Chou YL, Lin JR, Chen CH, Liu CH. Revealing a novel Decorin-expressing tumor stromal subset in hepatocellular carcinoma via integrative analysis single-cell RNA sequencing. Cancer Cell Int 2025; 25:194. [PMID: 40420150 DOI: 10.1186/s12935-025-03811-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality, emphasizing the need for novel therapeutic strategies. Decorin (DCN), a chondroitin sulfate proteoglycan (CSPG), has been proposed as a tumor suppressor, yet its precise role in HCC and the tumor microenvironment (TME) remains underexplored. Through integrated analyses of bulk RNA and single-cell RNA sequencing datasets, we identified a distinct tumor stromal subset highly expressing DCN and associated chondroitin sulfate (CS) synthases. Our findings revealed that DCN expression is significantly downregulated in HCC tissue, but upregulated in peri-tumor stroma, where it correlates with better prognosis and reduced capsular invasion. Western blot analysis demonstrated that CS-DCN, the glycosylated form of DCN, plays a dominant role in this context. Single-cell clustering analysis identified a unique stromal subset in HCC characterized by elevated expression of DCN, CSPGs, and CS synthases, associated with extracellular matrix (ECM) remodeling and protective barrier functions. A six-gene DCN-associated signature derived from this subset, including DCN, BGN, SRPX, CHSY3, CHST3, and CHPF, was validated as a prognostic marker for HCC. Furthermore, functional assays demonstrated that CS-DCN significantly inhibited HCC cell proliferation and invasion. Our study highlights the critical role of DCN in HCC TME and provides insights into its therapeutic potential. Modulating CSPG pathways, particularly on CS-DCN-expressing stromal cells, may offer a promising approach for improving HCC treatment and patient outcomes.
Collapse
Affiliation(s)
- Chi Hsiao
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
- Department of Pathology, School of Medicine, Chung Shan Medical University, Taichung, Republic of China
| | - Wen-Chieh Liao
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
| | - Ju-Pi Li
- Department of Pathology, School of Medicine, Chung Shan Medical University, Taichung, Republic of China
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Republic of China
| | - Yu-Cheng Chou
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Republic of China
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Yu-Lun Chou
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
| | - Jeng-Rong Lin
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
| | - Chia-Hua Chen
- Department of Anatomy, School of Medicine, Chang Gung University, Taoyuan, Republic of China.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Republic of China.
- Neuroscience Research Center, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Republic of China.
| | - Chiung-Hui Liu
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China.
| |
Collapse
|
7
|
Dong Z, Chang X, Luo X, Li H, Deng M, Huang Z, Chen T, Chen Y, Sun B, Wu Y, Wu R, Wu Q, Zhu J, Zheng J. Integration of network pharmacology, transcriptomics, and experimental verification to investigate the mechanism of action of cepharanthine hydrochloride against prostate cancer. Sci Rep 2025; 15:18115. [PMID: 40413339 PMCID: PMC12103559 DOI: 10.1038/s41598-025-03004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/20/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
The incidence of prostate cancer (PCa) is high among elderly men. Cepharanthine hydrochloride (CH) is recognized for its important role in the prevention and treatment of various diseases. However, its effects and mechanisms of action in the context of PCa remain unclear. Our study aims to examine the therapeutic role and mechanisms of action of CH in PCa. Targets of CH and PCa-related genes were identified using different databases, and the biological processes through which CH might exert its therapeutic effects were predicted via protein-protein interaction (PPI) network and enrichment analyses. Subsequently, the PCa cell lines PC-3 and DU145 were used to assess the concentration- and time-dependent effects of CH on cell viability, proliferation, and migration. Transcriptomic sequencing and differential expression analysis were used to identify the key target protein of CH and the key signaling pathways involved in its therapeutic effects against PCa. Molecular docking was used to analyze the binding between CH and its target protein. Additionally, quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blotting, gene knockout, pharmacological intervention, and tumor formation experiments were performed to validate the therapeutic effects and mechanisms of action of CH against PCa in vitro and in vivo. Network pharmacology showed that CH, a Chinese herbal medication, might prevent PCa by regulating protein phosphorylation-related biological processes. In vitro experiments showed that CH inhibited the proliferation and migration of PCa cells in a concentration-dependent manner. In addition, integration of transcriptomic sequencing, differential expression analysis, and GO enrichment analysis suggested that the ERK protein played a crucial role in the anti-tumor activity of CH. Molecular docking and molecular dynamics simulations revealed strong binding affinities between CH and ERK1/2. Further experimental verification, involving qRT-PCR, western blotting, gene knockout, pharmacological intervention, and tumor formation experiments, demonstrated that CH upregulated dual-specificity phosphatase (DUSP) 1 and suppressed the phosphorylation of ERK, thereby inhibiting the development and progression of PCa in vivo and in vitro. In conclusion, the findings of this study suggest that CH suppresses the ERK signaling pathway by enhancing the expression of DUSP1, thereby exerting anti-tumor effects against PCa in vitro and in vivo. Therefore, CH may serve as a novel therapeutic agent for PCa, showing remarkable potential for clinical application.
Collapse
Affiliation(s)
- Zongming Dong
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Xiaosa Chang
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Xing Luo
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Hui Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Ming Deng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Zeyu Huang
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Tingting Chen
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yu Chen
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Bishao Sun
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yingbing Wu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Ronghua Wu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Qingjian Wu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Jingzhen Zhu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400042, China.
| |
Collapse
|
8
|
Abushikha MAA, Karagac MS, Yesilkent EN, Ceylan H. Investigation of the effects of monosodium glutamate and tannic acid on the glutathione and thioredoxin systems in the liver of rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04279-5. [PMID: 40397121 DOI: 10.1007/s00210-025-04279-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
While there is no conclusive evidence that monosodium glutamate (MSG, a food additive) directly causes liver cancer in humans, certain studies suggest a potential link between MSG-induced liver injury and cancer development. This study aimed to evaluate the protective effect of tannic acid (TA, a natural polyphenol) against MSG-induced hepatotoxicity through the glutathione and thioredoxin systems. Twenty-four rats were randomly divided into control and experimental groups and treated with TA, MSG, and MSG+TA once daily by oral gavage for 21 days. In addition to major oxidative stress indicators (total glutathione; GSH + GSSG and malondialdehyde; MDA), mRNA expression changes and biological activity responses of components of the glutathione and thioredoxin systems were examined in the liver tissues of all animals. The results showed that MSG alone negatively affected both stress indicators and antioxidant system components (glutathione peroxidase; GPx, glutathione reductase; GR, glutathione-S-transferase; GST, and thioredoxin reductase; TrxR) in terms of mRNA expression and biological activity. However, the combination of MSG and TA demonstrated robust antioxidative effects, surpassing the outcomes of MSG treatment. Our results provide new insights into pivotal molecular targets and protective candidates that should be focused on in future in vivo and in vitro HCC research.
Collapse
Affiliation(s)
- Mohammad A A Abushikha
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, 25240, Erzurum, Türkiye
| | - Medine Sibel Karagac
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, 25240, Erzurum, Türkiye
| | - Esra Nur Yesilkent
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, 25240, Erzurum, Türkiye
| | - Hamid Ceylan
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, 25240, Erzurum, Türkiye.
- East Anatolian High Technology Research and Application Center (DAYTAM), Atatürk University, 25240, Erzurum, Türkiye.
| |
Collapse
|
9
|
Özbek M, Beyaz F, Öztop M, Karaca H, Cabir A, Kiryar BF. Anatolian ground squirrel (Spermophilus xanthoprymnus) retina: Comparative expression of synaptophysin, NeuN, calbindin-D28k, parvalbumin, glial fibrillary acidic protein, and Iba-1 during pre-hibernation and hibernation. Anat Rec (Hoboken) 2025. [PMID: 40377082 DOI: 10.1002/ar.25682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/18/2025]
Abstract
Hibernation induces significant molecular and cellular adaptations in the retina to maintain function under reduced metabolic conditions. This study aimed to investigate the expression of neuronal, synaptic, and glial markers in the retina of Spermophilus xanthoprymnus during pre-hibernation and hibernation periods using immunohistochemical staining. Synaptophysin expression, restricted to the inner plexiform layer (IPL) and outer plexiform layer (OPL) during pre-hibernation, significantly increased in both layers during hibernation, with additional expression observed in the outer nuclear layer. NeuN immunoreactivity remained unchanged in the ganglion cell layer (GCL) but increased notably in the INL during hibernation. Calbindin-D28k expression, prominent in the INL and plexiform layers during pre-hibernation, decreased markedly in hibernation. In contrast, parvalbumin expression increased across all retinal layers, except the photoreceptor layer, during hibernation. Glial fibrillary acidic protein (GFAP) expression, observed in the NFL and GCL, was significantly reduced during hibernation. Iba-1 immunoreactivity, sparse in the IPL and OPL during pre-hibernation, showed a pronounced increase in the IPL, OPL, and INL during hibernation periods. In conclusion, the expression of synaptophysin, NeuN, calbindin-D28k, parvalbumin, GFAP, and Iba-1 was investigated for the first time in the retina of the Anatolian ground squirrel during pre-hibernation and hibernation. This study reveals region-specific shifts in retinal marker expression during pre-hibernation and hibernation, providing a basis for future research into visual system adaptations and retinal plasticity under metabolic suppression.
Collapse
Affiliation(s)
- Mehmet Özbek
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Feyzullah Beyaz
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
- Department of Neuroscience, Gevher Nesibe Genome and Stem Cell Institute, Erciyes University, Kayseri, Turkey
| | - Mustafa Öztop
- Department of Biology, Faculty of Arts and Sciences, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Harun Karaca
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Ahmet Cabir
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Begüm Fatma Kiryar
- Department of Neuroscience, Gevher Nesibe Genome and Stem Cell Institute, Erciyes University, Kayseri, Turkey
| |
Collapse
|
10
|
Alzarea EA, Al-Kuraishy HM, Al-Gareeb AI, Alexiou A, Papadakis M, Beshay ON, Batiha GES. The Conceivable Role of Metabolic Syndrome in the Pathogenesis of Alzheimer's Disease: Cellular and Subcellular Alterations in Underpinning a Tale of Two. Neuromolecular Med 2025; 27:35. [PMID: 40379890 DOI: 10.1007/s12017-025-08832-6] [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: 10/31/2024] [Accepted: 01/09/2025] [Indexed: 05/19/2025]
Abstract
Alzheimer's disease (AD)is an age-related neurodegenerative disease characterized by memory decline and cognitive impairment .AD is common in people aged > 65 years, though most of AD cases are sporadic, which accounts for 95%, and 1-5% of AD is caused by familial causes . The causes of AD are aging, environmental toxins, and cardiometabolic factors that induce the degeneration of cholinergic neurons. It has been shown that the metabolic syndrome which is a clustering of dissimilar constituents including insulin resistance (IR), glucose intolerance, visceral obesity, hypertension, and dyslipidemia is implicated in the pathogenesis of AD. Metabolic syndrome disapprovingly affects cognitive function and the development in AD by inducing the development of oxidative stress, neuroinflammation, and brain IR. These changes, together with brain IR, impair cerebrovascular reactivity causing cognitive impairment and dementia. Nevertheless, the fundamental mechanism by which metabolic syndrome persuades AD risk is not entirely explicated. Accordingly, this review aims to discuss the connotation between metabolic syndrome and AD. In conclusion, metabolic syndrome is regarded as a possible risk factor for the initiation of AD neuropathology by diverse signaling pathways such as brain IR, activation of inflammatory signaling pathways, neuroinflammation, defective proteostasis, and dysregulation of lipid mediators.
Collapse
Affiliation(s)
- Ekremah A Alzarea
- Hematopathology, Department of Pathology, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu./Najaf-Iraq, PO.Box13, Kufa, Iraq
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, Australia
- Department of Research & Development, Funogen, Athens, Greece
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Olivia N Beshay
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
11
|
Sang W, Zhang X, Hu Q, Jiang B, Guan J, Huang Z, Sun L, Sun D. Inhibition of Dectin-1 alleviates inflammation in early diabetic retinopathy by regulating microglia phenotype. Gene 2025:149572. [PMID: 40381970 DOI: 10.1016/j.gene.2025.149572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/25/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Diabetic retinopathy (DR) is a major factor in vision loss in diabetic patients, triggering a series of pathological changes. At present, the treatment methods for diabetic retinopathy are limited. There is an urgent need to further explore its mechanism to bring more treatment options to patients.There is increasing evidence that microglia activation plays a crucial role in inflammatory DR. The C type lectin receptor Dectin-1 is known to play an important role in the inflammatory regulation of microglia, however, its role and mechanism in DR remains unclear. This study aims to elucidate the possible mechanisms through which Dectin-1 influences the inflammatory response in high glucose(HG) stimulated microglia and its impact on retinal inflammation during the early stages of DR. METHODS Human microglial cells (HMC3) were stimulated with HG (25 mmol/L), and a streptozotocin (STZ)induced C57BL/6J mouse model was established to simulate DR. To investigate the role of Dectin-1 in HMC3 cells and its underlying molecular mechanisms, we employed western blotting, quantitative realtime PCR (qRT-PCR), hematoxylineosin (H&E) staining, and immunofluorescence analysis. RESULTS Our findings revealed that Dectin-1 levels were elevated in microglia stimulated by HG, playing a pivotal role in cell polarization and the induction of inflammatory factors in vitro. In vivo experiments conducted on STZ induced diabetic mice demonstrated an increased expression of Dectin-1 in retinal tissues. This elevation further promoted the expression of pro inflammatory factors, such as TNF-α, IL-1β, and iNOS, triggering an inflammatory response and causing damage to the retina. Notably, inhibiting Dectin-1 reversed these detrimental effects, ultimately contributing to the delay in the progression of DR. Our investigation also uncovered a significant interaction between Dectin-1 and the downstream pro-inflammatory pathway NF-κB. This interaction occurred through the activation of spleen tyrosine kinase (Syk), both in vitro and in vivo. CONCLUSIONS In summary, our research strongly suggests that Dectin-1 plays a crucial pro-inflammatory role in early DR. This mechanismis, at least in part, mediated through the Syk/NF-κB pathway. Consequently, inhibition of Dectin-1 is expected to become a potential therapeutic target for delaying DR.
Collapse
Affiliation(s)
- Wei Sang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Future Medical Labotary, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Department of Ophthalmology, Qiqihar Eye & ENT Hospital, Qiqihaer 161000, China
| | - Xue Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Future Medical Labotary, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Qiang Hu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Future Medical Labotary, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Bo Jiang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jitian Guan
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Future Medical Labotary, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Zhangxin Huang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Future Medical Labotary, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Lijun Sun
- Department of Ophthalmology, Qiqihar Eye & ENT Hospital, Qiqihaer 161000, China
| | - Dawei Sun
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Future Medical Labotary, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
12
|
Pu Q, Gao F, Xiao Y, Wu J, Wang C, Mo X, Zhang Z, Zheng R, Wu D. Tobacco smoking exposure-mediated ELAVL1 regulates bladder cancer cell senescence via autophagy activation. Toxicology 2025; 516:154193. [PMID: 40378909 DOI: 10.1016/j.tox.2025.154193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Tobacco smoking is a well-established risk factor for bladder cancer, which shows connection to cell senescence in various diseases. However, the regulatory mechanisms linking tobacco smoking exposure to senescence regulation in bladder cancer remain incompletely characterized. In this investigation, we demonstrated that the smoking carcinogen 4-aminobiphenyl (4-ABP) inhibited cell senescence while enhancing proliferative, invasive, and migratory capacities of bladder cancer cells, as evidenced by SA-β-gal staining, western blot and cell malignant phenotype experiments. We further identified 275 cell senescence-related genes specific to bladder cancer based on CellAge database, the Nanjing bladder cancer dataset and public database. Through genome-wide association studies in 580 bladder cancer cases and 1101 controls, we pinpointed that rs12978895 G>A in ELAVL1 was significantly correlated with decreased bladder cancer risk (odds ratio = 0.79, 95 % confidence interval = 0.68-0.92) and interacted with smoking (P = 0.043). In genetic regulation, both experimental and population study showed that the A allele of rs12978895 significantly reduced ELAVL1 expression, while elevated ELAVL1 levels were observed in tumor tissues. Notably, exposed to smoking carcinogen 4-ABP resulted in a markedly increased expression of ELAVL1, which inhibited senescence of bladder cancer cells. Mechanistically, 4-ABP upregulated ELAVL1 suppressed cell senescence through autophagy activation, thus promoting bladder cancer progression. This study elucidated the genetic susceptibility and biological function of ELAVL1 in tobacco smoking exposure cell models, shedding light on the etiology of bladder cancer.
Collapse
Affiliation(s)
- Qiuyi Pu
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health; Institute of Clinical Research, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Fang Gao
- Department of Urology, The Third Affiliated Hospital of Nanjing Medical University (Changzhou Second People's Hospital), Changzhou, China
| | - Yanping Xiao
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health; Institute of Clinical Research, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Jiajin Wu
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health; Institute of Clinical Research, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, School of Public Health, Southeast University, Nanjing, China
| | - Chao Wang
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health; Institute of Clinical Research, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaoxiao Mo
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health; Institute of Clinical Research, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Zhengdong Zhang
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health; Institute of Clinical Research, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, China; Department of Occupational Medicine and Environmental Health, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, China.
| | - Rui Zheng
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health; Institute of Clinical Research, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, China; Department of Occupational Medicine and Environmental Health, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, China.
| | - Dongmei Wu
- Departments of Environmental Genomics and Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health; Institute of Clinical Research, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, China; Department of Occupational Medicine and Environmental Health, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, China.
| |
Collapse
|
13
|
Peter‐Okaka U, Boison D. Adenosine Kinase: An Epigenetic Modulator and Drug Target. J Inherit Metab Dis 2025; 48:e70033. [PMID: 40393929 PMCID: PMC12092209 DOI: 10.1002/jimd.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/28/2025] [Accepted: 04/15/2025] [Indexed: 05/22/2025]
Abstract
Adenosine kinase (ADK, EC: 2.7.1.20) is an evolutionarily ancient ribokinase, which acts as a metabolic regulator by transferring a phosphoryl group to adenosine to form AMP. The enzyme is of interest as a therapeutic target because its inhibition is one of the most effective means to raise the levels of adenosine and hence adenosine receptor activation. For these reasons, ADK has received significant attention in drug discovery efforts in the early 2000s for indications such as epilepsy, chronic pain, and inflammation; however, the report of adverse events regarding cardiovascular and hepatic function as well as instances of microhemorrhage in the brain of preclinical models prevented further development efforts. Recent findings emphasize the importance of compartmentalization of the adenosine system reflected by two distinct isoforms of the enzyme, ADK-S and ADK-L, expressed in the cytoplasm and the cell nucleus, respectively. Newly identified adenosine receptor independent functions of adenosine as a regulator of biochemical transmethylation reactions, which include DNA and histone methylation, identify ADK-L as a distinct therapeutic target for the regulation of the nuclear methylome. This newly recognized role of ADK-L as an epigenetic regulator points toward the potential disease-modifying properties of the next generation of ADK inhibitors. Continued efforts to develop therapeutic strategies to separate nuclear from extracellular functions of adenosine would enable the development of targeted therapeutics with reduced adverse event potential. This review will summarize recent advances in the discovery of novel ADK inhibitors and discuss their potential therapeutic use in conditions ranging from epilepsy to cancer.
Collapse
Affiliation(s)
- Uchenna Peter‐Okaka
- Department of NeurosurgeryRutgers New Jersey Medical School and Robert Wood Johnson Barnabas HealthNew BrunswickNew JerseyUSA
| | - Detlev Boison
- Department of NeurosurgeryRobert Wood Johnson and New Jersey Medical Schools, Rutgers HealthPiscatawayNew JerseyUSA
- Brain Health InstituteRutgers UniversityPiscatawayNew JerseyUSA
- Rutgers HealthRutgers Cancer InstituteNew BrunswickNew JerseyUSA
| |
Collapse
|
14
|
Oska N, Awad AM, Eltanani S, Shawky M, Naghdi A, Yumnamcha T, Singh LP, Ibrahim AS. Glyceraldehyde-3-phosphate dehydrogenase/1,3-bisphosphoglycerate-NADH as key determinants in controlling human retinal endothelial cellular functions: Insights from glycolytic screening. J Biol Chem 2025; 301:108472. [PMID: 40158853 PMCID: PMC12136781 DOI: 10.1016/j.jbc.2025.108472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025] Open
Abstract
Maintaining barrier integrity, along with cell adhesion to the extracellular matrix and the subsequent process of cell spreading, are essential functions of endothelial cells, including human retinal endothelial cells (HRECs). Disruptions in these processes can lead to vision-threatening conditions like diabetic retinopathy. However, the bioenergetic mechanisms that regulate HREC barrier function and cell spreading remain incompletely understood. This study investigates the role of lower glycolytic components in modulating these critical functions of HRECs. In vitro, Electric Cell-Substrate Impedance Sensing (ECIS) technology was used to measure real-time changes in HREC barrier integrity (electrical resistance) and cell spreading (capacitance). Pharmacological inhibitors targeting lower glycolytic components were tested: heptelidic acid for glyceraldehyde-3-phosphate dehydrogenase (GAPDH), NG-52 for phosphoglycerate kinase (PGK), shikonin for pyruvate kinase M (PKM), galloflavin for lactate dehydrogenase (LDH), AZD3965 for lactate transporter (MCT1), and MSDC-0160 for the mitochondrial pyruvate carrier (MPC). GAPDH knockdown was performed using siRNA, and cell viability was assessed via LDH release assays. For in vivo studies, wild-type C57BL/6J mice received intravitreal injections of heptelidic acid, while control mice received the vehicle (dimethyl sulfoxide). Retinal vascular permeability was assessed by fluorescein angiography (FA) and retinal albumin leakage. The most significant decrease in electrical resistance and increase in capacitance of HRECs were observed following the dose-dependent inhibition of GAPDH and the resulting reduction in 1,3-bisphosphoglycerate (1,3-BPG) and NADH by heptelidic acid. LDH level analysis at 24 to 48 h post-treatment with heptelidic acid (1 and 10 μM) showed no significant difference compared to controls, indicating that the observed disruption of HREC functionality was not due to cell death. Supporting these findings, inhibition of downstream glycolytic steps that result in the accumulation of 1,3-BPG and NADH, such as treatment with NG-52 for PGK or shikonin for PKM, led to a significant increase in electrical resistance and a decrease in cell capacitance. Furthermore, GAPDH knockdown via siRNA also led to a significant decrease in cellular resistance in HRECs. In vivo, FA imaging demonstrated that intravitreal injection of heptelidic acid led to significant retinal vascular leakage, as further supported by increased albumin extravasation in treated eyes. Conversely, pharmacological inhibition of other lower glycolytic components, including LDH, MCT, and MPC, did not significantly alter HREC barrier function or spreading behavior. This study highlights the distinct roles of lower glycolytic components in regulating HREC functionality. GAPDH and its downstream products (1,3-BPG and NADH) are shown to play a pivotal role in maintaining barrier integrity and promoting HREC adhesion and spreading. These findings guide the development of targeted interventions that modulate HREC bioenergetics to treat endothelial dysfunction in various retinal disorders, while minimizing potential adverse effects on healthy endothelial cells.
Collapse
Affiliation(s)
- Nicole Oska
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Ahmed M Awad
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa, Egypt
| | - Shaimaa Eltanani
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Mohamed Shawky
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan, USA; Department of Biochemistry, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Armaan Naghdi
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Thangal Yumnamcha
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Lalit Pukhrambam Singh
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Ahmed S Ibrahim
- Department of Ophthalmology, Visual, and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan, USA; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan, USA; Molecular Therapeutics Research Program, Karmanos Cancer Institute (KCI), School of Medicine, Wayne State University, Detroit, Michigan, USA.
| |
Collapse
|
15
|
Bahriz HA, Abdelaziz RR, El-Kashef DH. Allopurinol abates hepatocellular carcinoma in rats via modulation of NLRP3 inflammasome and NF-κB pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6043-6058. [PMID: 39636403 DOI: 10.1007/s00210-024-03666-8] [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: 05/02/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
The present research was performed to examine the possible capability of allopurinol to prevent developing hepatocellular carcinoma (HCC) and to explore the fundamental mechanisms that control the hepatoprotective effect considering the enormous impact of HCC on patients' quality of life. Male Sprague Dawely rats were given i.p. injection of thioacetamide (TAA) (200 mg/kg) twice a week for 16 weeks in order to induce HCC. The histological analysis and assessment of the serum levels of liver function indicators verified the development of HCC. Two regimens of allopurinol (100 mg/kg, p.o.) were used; the first involved giving it concurrently with TAA from week 13 to week 16, and the second regimen started from week 9 to week 16. Chronic TAA damage was associated with considerable overexpression of the profibrogenic cytokine TGF-β, degradation and nuclear translocation of NF-κB, which released a number of inflammatory mediators, and upregulation of the NLRP3/caspase1 pathway. Administration of allopurinol demonstrated considerable enhancements in liver function and oxidative balance. Moreover, pathological characteristics like cirrhosis, dysplastic changes, and HCC nodules were greatly diminished. Allopurinol via suppressing TGF-β expression, inhibiting NF-κB nuclear translocation, and restricting inflammatory NLRP3/caspase1/IL-1β pathway was able to protect against TAA-induced liver damage, and it could be a promising therapy for HCC.
Collapse
MESH Headings
- Animals
- Allopurinol/pharmacology
- Allopurinol/therapeutic use
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Male
- NF-kappa B/metabolism
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/prevention & control
- Inflammasomes/metabolism
- Rats, Sprague-Dawley
- Thioacetamide
- Signal Transduction/drug effects
- Rats
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/chemically induced
- Liver Neoplasms/drug therapy
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/chemically induced
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/prevention & control
- Liver Neoplasms, Experimental/drug therapy
- Liver/drug effects
- Liver/pathology
- Liver/metabolism
Collapse
Affiliation(s)
- Heba A Bahriz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
16
|
El-Shiekh RA, Mohamed AF, Mandour AA, Adel IM, Atwa AM, Elgindy AM, Esmail MM, Senna MM, Ebid N, Mustafa AM. Hesperidin in Chronic Fatigue Syndrome: An Integrated Analysis of Traditional Pharmacology and Machine Learning-Based Therapeutic Predictions. Chem Biodivers 2025:e202403506. [PMID: 40234200 DOI: 10.1002/cbdv.202403506] [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: 01/04/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/17/2025]
Abstract
Hesperidin, a bioflavonoid abundantly found in citrus fruits, offers a myriad of health benefits. With the food industry extensively utilizing citrus fruits, particularly for juice production, substantial quantities of by-products such as peels, seeds, cells, and membrane residues accumulate. Remarkably, these by-products serve as a valuable source of hesperidin. Consequently, the extraction of hesperidin from these by-products has garnered significant scientific interest, aiming to harness its potential as a natural antioxidant. By shedding light on these aspects, this review provides a comprehensive review of hesperidin's role in enhancing human well-being, particularly in the context of chronic fatigue syndrome (CFS). By synthesizing current research, we elucidate the compound's antioxidant, anti-inflammatory, and neuroprotective effects, which may mitigate symptoms associated with CFS. Furthermore, we introduce machine learning methodologies to predict hesperidin's efficacy in clinical settings, offering a novel perspective on personalized nutrition strategies. Our findings underscore the need for further empirical studies to validate these predictions and explore hesperidin's mechanisms of action. This review not only bridges the gap between nutrition science and pharmacology but also highlights the promising future of hesperidin as a nutraceutical in combating chronic health conditions.
Collapse
Affiliation(s)
- Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Faculty of Pharmacy, King Salman International University (KSIU), South Sinai, Egypt
| | - Asmaa A Mandour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), Cairo, Egypt
| | - Islam M Adel
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed M Atwa
- College of Pharmacy, Al-Ayen Iraqi University, AUIQ, An Nasiriyah, Iraq
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Ali M Elgindy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Manar M Esmail
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Mohamed Magdy Senna
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Nouran Ebid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Aya M Mustafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| |
Collapse
|
17
|
Zhu Z, Lian X, Hu J, Wang Z, Zhong Y, Zhao Y, Lu L, Pan Y, Zhou M, Xu J. DPHC from Alpinia officinarum Hance specifically modulates the function of CENPU in the cell cycle and apoptosis to ameliorate hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119598. [PMID: 40058474 DOI: 10.1016/j.jep.2025.119598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alpinia officinarum Hance (A. officinarum), a perennial herb used in the treatment of digestive system cancers, holds significant value for the Li people of Hainan as a traditional Chinese medicine. (R)-5-hydroxy-1,7-diphenyl-3-heptanone (DPHC), a diarylheptanoid component is derived from A. officinarum. Diarylheptanoids have demonstrated anti-proliferative effects on breast cancer cells, neuroblastoma cells, and other tumor cells. However, the pharmacological activity of DPHC in improving hepatocellular carcinoma (HCC) remains undefined. AIM OF THE STUDY To elucidate the anti-HCC effects of DPHC derived from A. officinarum and explore its underlying mechanistic pathways both in vivo and in vitro. MATERIAL AND METHODS The effects of DPHC on HCC cell lines were evaluated in vitro using cell counting kit-8, EdU cell proliferation assays, a wound healing assay, a three-dimensional tumor spheroid model, and flow cytometry. The ability of DPHC to ameliorate HCC was assessed in vivo via a nude mouse subcutaneous xenograft tumor model, serum biochemical marker detection, and hematoxylin-eosin staining. The molecular mechanism of DPHC in HCC was elucidated through a combination of transcriptome sequencing, cell transfection, immunohistochemistry assay, immunofluorescence staining, quantitative reverse transcription-PCR, and western blot analysis. RESULTS DPHC induced significant G0/G1 phase arrest and apoptosis in HepG2 and HCCLM3 cells while also markedly inhibiting tumor growth in nude mice. Mechanically, DPHC directly interacted with centromere-associated protein U (CENPU) to suppress its expression. The reduced expression of CENPU results in decreased interaction with the transcription factor E2F6, thereby affecting the transcriptional activity of the transcription factor E2F1. This subsequently inhibits the expression of downstream cell cycle factors (CCND1, CDK4, and CDK1) and increases apoptosis factors (Caspase 3 and Caspase 9). CONCLUSIONS DPHC from A. officinarum specifically modulates the function of CENPU in the cell cycle and apoptosis to ameliorate HCC. Our study revealed the anti-HCC effect and underlying mechanism of DPHC, offering new insights and potential targets for HCC treatment.
Collapse
Affiliation(s)
- Zhe Zhu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Xiuxia Lian
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education & International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province & Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, Hainan, 571199, China
| | - Jicheng Hu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Zhe Wang
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Yinghong Zhong
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Yuan Zhao
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Lu Lu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Yipeng Pan
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China.
| | - Mingyan Zhou
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China.
| | - Jian Xu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China; Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
18
|
Uslu H, Atila Uslu G, Çiçek B, Bolat İ, Yıldırım S. Trigonelline alkaloid is effective in preventing doxorubicin-induced lung damage. Arch Physiol Biochem 2025; 131:169-176. [PMID: 39287053 DOI: 10.1080/13813455.2024.2404097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/21/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND One of the most popular chemotherapy medications is doxorubicin (DOX), however it can have non-negligible damage. When the underlying mechanisms of damage are investigated, the most prominent pathways are oxidative stress, inflammation and apoptosis. AIM We investigated the NF-κB/MAPK inflammatory pathway and cellular apoptosis to determine the efficacy of trigonelline alkaloid (TRIG) in preventing DOX-induced lung injury. METHODOLOGY The study consisted of C, TRIG, DOX and TRIG+DOX groups. TRIG and TRIG+DOX groups received 50 mg/kg TRIG for 7 days. On day 8, DOX and TRIG+DOX groups received a single dose of 15 mg/kg DOX. RESULTS Our results showed that apoptosis markers and inflammation were higher in the DOX group. In contrast, TRIG pretreatment partially suppressed apoptosis and decreased inflammation by blocking the activation of the MAPK/NF-κB pathway, lowering IL-6 levels, and protecting the lung from apoptotic cell death. CONCLUSION Assessing TRIG's effectiveness in lung tissue injury, this study may be a crucial first step.
Collapse
Affiliation(s)
- Hamit Uslu
- Department of Physiology, Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Gözde Atila Uslu
- Department of Physiology, Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Betül Çiçek
- Department of Physiology, Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - İsmail Bolat
- Department of Pathology, Atatürk University, Erzurum, Türkiye
| | - Serkan Yıldırım
- Department of Pathology, Atatürk University, Erzurum, Türkiye
| |
Collapse
|
19
|
Wu X, Xiao X, Su Y, Zhang Y, Li G, Wang F, Du Q, Yang H. Use quercetin for pulmonary fibrosis: a preclinical systematic review and meta-analysis. Inflammopharmacology 2025; 33:1879-1897. [PMID: 40038212 DOI: 10.1007/s10787-025-01678-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is an age-related interstitial lung disease, which lacks effective drug treatment at present. Quercetin has been shown to have favorable anti-inflammatory and anti-fibrotic properties, and preliminary evidence suggests its potential efficacy and tolerability in PF patients. However, a comprehensive systematic review and evaluation of the protective effects and potential mechanisms of quercetin in PF models remains to be completed. Therefore, we conducted this study. METHODS The PubMed, Cochrane Library, Embase, and Web of Science databases were searched up to the April 1, 2024. CAMARADES was the methodological quality assessment tool. And statistical analyses were conducted with R and Stata 16.0. Origin was used for a three-dimensional (3D) dosage-intervention duration-efficacy model for quercetin treatment of PF. RESULTS A total of 20 studies, encompassing 44 independent experiments and involving 1019 animals, were included in the analysis. Meta-analysis revealed that quercetin significantly mitigated lung pathological tissue scores and the expression of lung fibrosis markers in PF animal models. Furthermore, quercetin significantly ameliorated inflammatory responses, oxidative stress, epithelial-mesenchymal transition and myofibroblast activation, cell senescence and apoptosis, and the markers expression of extracellular matrix (ECM) deposition. Quercetin did not show significant hepatic and nephrotoxicity. The 3D dosage-intervention duration-efficacy model indicated that a dosing period over 20 days and dosages range of 5-100 mg/kg were appropriate modalities. CONCLUSION Herein, our study highlights the potential of quercetin in the treatment of PF and the available mechanisms.
Collapse
Affiliation(s)
- Xuanyu Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xiang Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yuchen Su
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yuwei Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Ganggang Li
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Fei Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Quanyu Du
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Han Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| |
Collapse
|
20
|
Guyot E. Heparan sulfate chains in hepatocellular carcinoma. Gastroenterol Rep (Oxf) 2025; 13:goaf023. [PMID: 40093586 PMCID: PMC11908768 DOI: 10.1093/gastro/goaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 11/13/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Hepatocellular carcinoma (HCC) corresponds to the vast majority of liver cancer cases, with one of the highest mortality rates. Major advances have been made in this field both in the characterization of the molecular pathogenesis and in the development of systemic therapies. Despite these achievements, biomarkers and more efficient treatments are still needed to improve its management. Heparan sulfate (HS) chains are polysaccharides that are present at the cell surface or in the extracellular matrix that are able to bind various types of molecules, such as soluble factors, affecting their availability and thus their effects, or to contribute to interactions that position cells in their environments. Enzymes can modify HS chains after their synthesis, thus changing their properties. Numerous studies have shown HS-related proteins to be key actors that are associated with cellular effects, such as tumor growth, invasion, and metastasis, including in the context of liver carcinogenesis. The aim of this review is to provide a comprehensive overview of the biology of HS chains and their potential importance in HCC, from biological considerations to clinical development, and the identification of biomarkers, as well as therapeutic perspectives.
Collapse
Affiliation(s)
- Erwan Guyot
- Biochemistry Unit, Saint-Antoine Hospital, AP-HP Sorbonne University, Paris Cedex, France
| |
Collapse
|
21
|
Burman MD, Bag S, Ghosal S, Bhowmik S. Glycation of Proteins and Its End Products: From Initiation to Natural Product-Based Therapeutic Preventions. ACS Pharmacol Transl Sci 2025; 8:636-653. [PMID: 40109756 PMCID: PMC11915047 DOI: 10.1021/acsptsci.4c00684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Diabetes is a chronic metabolic disorder characterized by elevated blood glucose levels, which lead to the glycation of proteins and the formation of advanced glycation end products (AGEs). These AGEs contribute to oxidative stress, inflammation, and the development of complications such as cardiovascular disease, nephropathy, and anemia, significantly increasing mortality rates among diabetic patients. This Review focuses on the role of glycation inhibitors as a potential strategy to prevent AGE-related pathologies. While synthetic glycation inhibitors have shown promise, their adverse effects highlight the need for safer alternatives. We specifically explore a range of natural compounds-flavonoids, curcuminoids, terpenes, stilbenes, lignans, and coumarins-that have demonstrated significant antiglycating properties. The mechanisms through which these natural products inhibit glycation, including antioxidant activity, metal ion chelation, and direct interference with the glycation process, are discussed in detail. This review underscores the potential of natural products as effective and safer glycation inhibitors, offering a promising avenue for the development of therapeutic strategies against diabetes and AGE-related disorders.
Collapse
Affiliation(s)
- Mangal Deep Burman
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
| | - Sagar Bag
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
| | - Souvik Ghosal
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Pondy-Cuddalore Main Road, Pillaiyarkuppam, Pondicherry 607402, India
| | - Sudipta Bhowmik
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Pondy-Cuddalore Main Road, Pillaiyarkuppam, Pondicherry 607402, India
| |
Collapse
|
22
|
Huang R, Tang X, Liu S, Sun L. Decoding CKD-induced muscle atrophy through the critical role of lncRNA GAS5 and pyroptosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102451. [PMID: 39974290 PMCID: PMC11835621 DOI: 10.1016/j.omtn.2025.102451] [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: 08/03/2024] [Accepted: 01/10/2025] [Indexed: 02/21/2025]
Abstract
Skeletal muscle atrophy is a prevalent complication of chronic kidney disease (CKD) and serves as an indicator of adverse prognosis and poor quality of life; however, the underlying mechanisms remain ambiguous. Emerging evidence has shown that long non-coding RNAs (lncRNAs) are involved in the pathogenesis of skeletal muscle atrophy. Using RNA sequencing (RNA-seq), we discerned elevated GAS5 expression in the muscles of CKD mice and verified these findings by real-time qPCR. Transmission electron microscopy confirmed morphological signs of pyroptosis, a potentially causal cellular death form. Additionally, elevated levels of pyroptosis markers, such as NLRP3, cleaved caspase-1, and GSDMD-N, were observed in CKD mouse models and lipopolysaccharide (LPS)/ATP-stimulated C2C12 myotubes. Intriguingly, the knockdown of GAS5 reduced these markers, alleviating pyroptosis and enhancing myofiber size, both in vitro and in vivo. Furthermore, we pinpointed an interaction between GAS5 and the mitochondrial translation elongation factor (TUFM) through RNA pull-down and mass spectrometry. This interaction amplified NLRP3 activity, contributing to pyroptosis and muscle atrophy. Notably, overexpressing TUFM counterbalanced this effect. Fundamentally, the interaction between GAS5 and TUFM appears to compromise the anti-pyroptosis capacity of TUFM. Consequently, this amplifies the activation of the NLRP3 pathway, which may underpin the crucial mechanism driving pyroptosis-mediated muscle atrophy. Our findings provide new evidence for GAS5's role in regulating cellular pyroptosis in CKD-induced skeletal muscle atrophy.
Collapse
Affiliation(s)
- Rong Huang
- Department of Nephrology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xinying Tang
- Department of Nephrology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Shuang Liu
- Department of Nephrology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Lijing Sun
- Department of Nephrology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
23
|
Rezaei H, Ravankhah M, Ansari M, Alirezaee A, Keshavarzian O, Abdollahi M, Sabet HR. Effects of Alpha-Lipoic Acid Supplementation on Weight Loss, Inflammatory, Lipid, and Hematological Levels in Patients With Chronic Kidney Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Ren Nutr 2025; 35:289-299. [PMID: 39413860 DOI: 10.1053/j.jrn.2024.08.004] [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: 11/12/2023] [Revised: 07/21/2024] [Accepted: 08/04/2024] [Indexed: 10/18/2024] Open
Abstract
OBJECTIVES The effects of alpha-lipoic acid (ALA) supplementation on cardiovascular-related factors have been evaluated in a number of randomized clinical trials, with different results. Thus, in this meta-analysis, the effects of ALA on blood levels of inflammatory, lipid, and hematological markers as well as anthropometric indices in patients with chronic kidney disease (CKD) were evaluated. METHODS Five electronic databases were used to conduct a comprehensive search through October 2023. Risk of bias assessment and data extraction were carried out separately by 2 reviewers on the included papers. The data were analyzed using the random-effects model in meta-analyses. The data were analyzed using the random-effects model in meta-analyses. We assessed inter-study heterogeneity with I2 and Cochran's Q test. RESULTS Nine of the 421 potential reports were included. Using random-effects models, no significant changes were observed in weight loss, body mass index, hemoglobin, and iron following ALA supplementation (600 mg/day). Results exhibited that ALA significantly reduced high-sensitivity C-reactive protein levels in individuals with CKD (weighted mean difference (WMD) = -2.91 mg/L, 95% CI: -4.65, -1.17, I2 = 50.5%, P = .09); however, there were no significant variations in levels of interleukin-6 (IL-6) or malondialdehyde. Regarding lipid profiles, findings revealed that ALA administration had no significant impact on high-density lipoprotein cholesterol and triglycerides levels among patients with CKD. However, compared to the control group, total cholestrol levels were considerably lower in CKD patients (WMD = -5.48 mg/dL, 95% CI: -10.55, -0.41, I2 = 0.0%, P = .50). Moreover, the sensitivity analyses showed that pooled WMDs for low-density lipoprotein cholesterol levels were significantly changed (-6.88 mg/dL, 95% CI, -12.78, -0.98). CONCLUSIONS These findings revealed that ALA supplementation slightly but significantly reduced blood levels of high-sensitivity C-reactive protein, total cholestrol, and low-density lipoprotein cholesterol, but did not affect IL-6, malondialdehyde, high-density lipoprotein cholesterol, weight, body mass index, iron, and hemoglobin in patients with CKD.
Collapse
Affiliation(s)
- Hadi Rezaei
- Department of Urology, Fars-Iranian Urological Association, Fars Society of Urology, Shiraz, Iran
| | - Mahdi Ravankhah
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Aida Alirezaee
- Student Research Committee, Mashad University of Medical Sciences, Mashhad, Iran
| | - Omid Keshavarzian
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhan Abdollahi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Reza Sabet
- Medical Journalism Department, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
24
|
Chen M, Song L, Zeng A. Harnessing nature's arsenal: Targeting the TGF-β/Smad Cascade with novel natural anti-fibrotic agents. Fitoterapia 2025; 181:106372. [PMID: 39778722 DOI: 10.1016/j.fitote.2024.106372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Hepatic fibrosis is a wound healing response that leads to excessive deposition of extracellular matrix (ECM) due to sustained liver injury. Hepatic stellate cells (HSCs) are key players in ECM synthesis, with the TGF-β/Smad signaling pathway being central to their activation. Despite advances in understanding the pathogenesis of hepatic fibrosis, effective anti-fibrotic therapies are still lacking. METHODS This treatise conducts a comprehensive review of the literature on the hepatoprotective effects of natural products, including natural medicine compounds, herbal extracts, and polysaccharides. The focus is on their ability to modulate the TGF-β pathway, which is critical in the activation of HSCs and ECM synthesis in hepatic fibrosis. RESULTS The review identifies a variety of natural products that have shown promise in inhibiting the TGF-β/Smad signaling cascade, thereby reducing the activation of HSCs and ECM accumulation. These findings highlight the potential of these natural products as therapeutic agents in the treatment of hepatic fibrosis. CONCLUSIONS The exploration of natural products as modulators of the TGF-β pathway presents a novel avenue for both clinical and preclinical research into hepatic fibrosis. Further investigation is warranted to fully understand the mechanisms of action and to develop these compounds into effective anti-fibrotic pharmaceuticals.
Collapse
Affiliation(s)
- Maohua Chen
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine,Chengdu, Sichuan 610041, PR China; Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China. Chengdu, Sichuan 610072, PR China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China.
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine,Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
25
|
Guo W, Yang Y, Liu G, Zhao J, Zhang Y, Li Y, Yang B, Zhu X, Li D, Qin X, Zhang P, Yang Z, Guo W, Kong D, Zhang W. The anti-neuroinflammatory effects of cepharanthine in uric acid-induced neuroinflammation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119409. [PMID: 39870338 DOI: 10.1016/j.jep.2025.119409] [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: 06/25/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cepharanthine (CEP) is an alkaloid extracted from Stephania cephalantha Hayata, a traditional Chinese medicine (TCM) renowned for its heatclearing and dehumidifying properties. For centuries, Stephania cephalantha Hayata has been employed in the treatment of a wide range of diseases, including pain, edema, inflammation, and fever. AIM OF THE STUDY Our research aims to investigate the role and mechanism of Cepharanthine in ameliorating uric acid (UA) induced neuroinflammatory responses. MATERIALS AND METHODS The Connectivity Map (CMap) was utilized to identify the therapeutic drug Cepharanthine, based on the proteomic disturbances associated with uric acid (UA). Limited proteolysis small molecule mapping (LiP-SMap) and thermal proteome profiling (TPP) technologies were used to identify the direct target proteins for UA and Cepharanthine. Additionally, we used the induced-fit docking algorithm integrated within the Schrodinger suite to explore the interactions between Cepharanthine and uric acid targets. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) technology was employed to determine the concentration of Cepharanthine in the mice cerebral cortex. The pro-inflammatory cytokine genes were also quantified by qPCR in U251 cells and in hyperuricemic mice. RESULTS The findings indicated that uric acid increased the transcription of pro-inflammatory cytokines and the expression levels of proteins linked to inflammation in U251 cells. PPP2R1A was identified as a potential candidate for direct interaction with uric acid, potentially initiating inflammation. Based on the CMap prediction, Cepharanthine was identified as a candidate drug for interaction with PPP2R1A. TPP analysis indicated that Cepharanthine could reduce the thermal stability of PPP2R1A. Molecular docking confirmed that Cepharanthine could directly bind to PPP2R1A. Furthermore, the detection of Cepharanthine in the cerebral cortex suggested its ability to cross the blood-brain barrier. Proteomic analysis of Cepharanthine-treated mice revealed significant enrichments of differentially expressed proteins (DEPs) in inflammation-related pathways and biological processes. Additionally, Cepharanthine was effective in decreasing the expression of pro-inflammatory cytokine genes induced by uric acid in U251 cells and in hyperuricemic mice. CONCLUSION Cepharanthine could effectively alleviate hyperuricemia-induced neuroinflammation via binding to PPP2R1A. This study offers a novel approach for prevention and treatment of neurological diseases caused by hyperuricemia.
Collapse
Affiliation(s)
- Wenyan Guo
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China; The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, 050017, China
| | - Yi Yang
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China; The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Guangyuan Liu
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Jiaojiao Zhao
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Yuyu Zhang
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Yahui Li
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Bingkun Yang
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China; Department of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Xiaoque Zhu
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China; CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang, Hebei Province, 050017, China
| | - Dandan Li
- Department of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China; CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang, Hebei Province, 050017, China
| | - Xia Qin
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Panpan Zhang
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Zuxiao Yang
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Wei Guo
- Department of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Dezhi Kong
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China.
| | - Wei Zhang
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, The Key Laboratory of Tranquilizing TCM, Hebei Provincial Administration of Traditional Chinese Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China.
| |
Collapse
|
26
|
Zhao H, Wang Q, Yang L, Ran Y, Hu Q, Hong Y, Tian M. Phytochemical analysis, antioxidant, anti-inflammatory and enzyme inhibitory activities of bean pear ( Pyrus calleryana fruit). FRONTIERS IN PLANT SCIENCE 2025; 16:1521990. [PMID: 39990718 PMCID: PMC11842399 DOI: 10.3389/fpls.2025.1521990] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/23/2025] [Indexed: 02/25/2025]
Abstract
Pyrus calleryana fruit (bean pear) is processed into fruit wine and used in traditional Chinese medicine. The present study reported phytochemical constituents, antioxidant, anti-inflammatory, and enzyme inhibitory activities of P. calleryana fruit water extract (WE) and ethanol extract (EE). In the P. calleryana fruit WE and EE, 63 compounds were identified using UHPLC-Q-Orbitrap-MS analysis, including 23 phenols, 13 flavonoids, 14 terpenoids, and 13 other types of compounds. In the antioxidant activity, WE and EE showed marked free radical scavenging effects on both ABTS (2.33 ± 0.15 μg/mL and 2.23 ± 0.15 μg/mL, respectively) and DPPH (5.93 ± 0.55 μg/mL and 7.07 ± 0.23 μg/mL, respectively), especially, their scavenging effects on DPPH free radicals were superior or equivalent to that of BHT (7.47 ± 0.47 μg/mL). In LPS-induced RAW264.7 cells, P. calleryana fruit WE and EE remarkably inhibited the secretion of inflammatory factors, and the inhibitory effect of WE on the release of IL-6, NO, and PGE2 was superior or equivalent to that of EE. Interestingly, P. calleryana fruit WE and EE exhibited potent inhibition on α-glucosidase (0.60 ± 0.09 μg/mL and 0.48 ± 0.09 μg/mL, respectively) and tyrosinase (210.11 ± 2.59 μg/mL and 45.35 ± 0.96 μg/mL, respectively), which were superior to their respective positive controls acarbose (302.57 ± 22.09 μg/mL) and arbutin (243.07 ± 15.91 μg/mL). Our findings suggested that P. calleryana fruit WE and EE possess significant antioxidant, anti-inflammatory, α-glucosidase, and tyrosinase inhibitory properties. Thus, P. calleryana fruit has great potential for application in functional food products.
Collapse
Affiliation(s)
- Huan Zhao
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang, China
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, China
| | - Qinqin Wang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Lanlan Yang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang, China
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, China
| | - Yuanquan Ran
- First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qiong Hu
- First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yi Hong
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Minyi Tian
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang, China
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, China
- First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
27
|
Xian J, Huang Y, Bai J, Liao Q, Chen Q, Cheng W, Su Z, Li S, Wu Y, Li J, Zhang J. Recent Advances in the Anti-Obesity Benefits of Phytoconstituents: From Phytochemistry to Targeting Novel-Systems. Phytother Res 2025; 39:630-660. [PMID: 39629748 DOI: 10.1002/ptr.8400] [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: 08/08/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 02/19/2025]
Abstract
Obesity is a metabolic disorder that has become a global health concern. The existing pharmaceutical drugs for treating obesity have some side effects. Compounds from natural sources are prospective substitutes for treating chronic diseases such as obesity, with the added advantages of being safe and cost-effective. However, due to factors such as poor solubility, low bioavailability, and instability in the physiological environment, the therapeutic efficacy of phytoconstituents is limited. Nowadays, developing nanoscaled systems has emerged as a vital strategy for enhancing the delivery and therapeutic effect of phytoconstituents. The present study discusses and categorizes phytoconstituents with anti-obesity effects and concludes the main mechanisms underlying their effects. Importantly, strategies used to develop phytoconstituent-based nano-drug delivery systems (NDDS) for obesity treatment that show improved efficacy relative to traditional administration routes are reviewed. Finally, the progress of research on phytoconstituent-based NDDS for obesity treatment is summarized to provide a reference for the development of safe and effective treatment strategies for obesity.
Collapse
Affiliation(s)
- Jing Xian
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinrong Bai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Liao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Qiyan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Weijian Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziye Su
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyao Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
28
|
Zhang H, Lu C, Yao Q, Jiao Q. In silico study to identify novel NEK7 inhibitors from natural sources by a combination strategy. Mol Divers 2025; 29:139-162. [PMID: 38598164 DOI: 10.1007/s11030-024-10838-4] [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: 09/02/2023] [Accepted: 03/06/2024] [Indexed: 04/11/2024]
Abstract
Cancer poses a significant global health challenge and significantly contributes to mortality. NEK7, related to the NIMA protein kinase family, plays a crucial role in spindle assembly and cell division. The dysregulation of NEK7 is closely linked to the onset and progression of various cancers, especially colon and breast cancer, making it a promising target for cancer therapy. Nevertheless, the shortage of high-quality NEK7 inhibitors highlights the need for new therapeutic strategies. In this study, we utilized a multidisciplinary approach, including virtual screening, molecular docking, pharmacokinetics, molecular dynamics simulations (MDs), and MM/PBSA calculations, to evaluate natural compounds as NEK7 inhibitors comprehensively. Through various docking strategies, we identified three natural compounds: (-)-balanol, digallic acid, and scutellarin. Molecular docking revealed significant interactions at residues such as GLU112 and ALA114, with docking scores of -15.054, -13.059, and -11.547 kcal/mol, respectively, highlighting their potential as NEK7 inhibitors. MDs confirmed the stability of these compounds at the NEK7-binding site. Hydrogen bond analysis during simulations revealed consistent interactions, supporting their strong binding capacity. MM/PBSA analysis identified other crucial amino acids contributing to binding affinity, including ILE20, VAL28, ILE75, LEU93, ALA94, LYS143, PHE148, LEU160, and THR161, crucial for stabilizing the complex. This research demonstrated that these compounds exceeded dabrafenib in binding energy, according to MM/PBSA calculations, underscoring their effectiveness as NEK7 inhibitors. ADME/T predictions showed lower oral toxicity for these compounds, suggesting their potential for further development. This study highlights the promise of these natural compounds as bases for creating more potent derivatives with significant biological activities, paving the way for future experimental validation.
Collapse
Affiliation(s)
- Heng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Chenhong Lu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Qilong Yao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Qingcai Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
29
|
Bahriz HA, Abdelaziz RR, El-Kashef DH. Desloratadine mitigates hepatocellular carcinoma in rats: Possible contribution of TLR4/MYD88/NF-κB pathway. Toxicol Appl Pharmacol 2025; 495:117202. [PMID: 39672344 DOI: 10.1016/j.taap.2024.117202] [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: 04/28/2024] [Revised: 07/03/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024]
Abstract
Chemotherapeutic medication-induced systemic toxicity makes cancer treatment less effective. Thus, the need for drug repurposing, which aids in the development of safe and efficient cancer therapies, is urgent. The primary goal of this research was to assess desloratadine hepatoprotective abilities and its capacity to attenuate TLR4/MyD88/NF-κB inflammatory pathway in hepatocellular carcinoma (HCC) induced by thioacetamide (TAA). Male Sprague Dawely rats received TAA injections (200 mg/kg, i.p., 2 times/week) for 16 weeks. To confirm the development of HCC, liver function biomarkers and histopathological analysis were evaluated. Desloratadine (5 mg/kg, p.o.) was administered to rats in 2 treatment groups; HCC + DES 1 group received desloratadine with TAA for 1 month from week 13-16, HCC + DES 2 group received desloratadine with TAA for 2 months from week 9-16. Chronic TAA administration resulted in considerable overexpression of the profibrogenic cytokine TGF-β and elevation in protein expression of NF-κB besides levels of TLR4, MyD88, TRAF6, TAK1 and IL-1β. Desloratadine administration showed a significant improvement in liver function tests, as well as an increase in tissue antioxidant enzymes and an improvement in the liver's histopathological features. Collectively, desloratadine through modulating TLR4/MyD88/TRAF6/TAK1/NF-κB and acting as an antioxidant, is a promising treatment for HCC induced by TAA.
Collapse
MESH Headings
- Animals
- Toll-Like Receptor 4/metabolism
- Male
- Myeloid Differentiation Factor 88/metabolism
- Loratadine/analogs & derivatives
- Loratadine/pharmacology
- Loratadine/therapeutic use
- NF-kappa B/metabolism
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/prevention & control
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Rats
- Liver Neoplasms/chemically induced
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Thioacetamide
- Liver Neoplasms, Experimental/chemically induced
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/prevention & control
- Liver/drug effects
- Liver/pathology
- Liver/metabolism
Collapse
Affiliation(s)
- Heba A Bahriz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
30
|
Karras GI, Colombo G, Kravats AN. Hsp90: Bringing it all together. Cell Stress Chaperones 2025; 30:69-79. [PMID: 39889818 PMCID: PMC12013134 DOI: 10.1016/j.cstres.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025] Open
Abstract
Heat-shock protein 90 (Hsp90) is an ancient and multifaceted protein-folding machine essential for most organisms. The past 40 years have uncovered remarkable complexity in the regulation and function of Hsp90, which dwarfs most other machines in the cell in sophistication. Here, we propose four analogies to illustrate Hsp90's sophistication: a multifunctional Swiss Army knife, an automobile engine and its controls, a switchboard acting as a hub and directing signals, and an orchestra conductor setting the tempo of a symphony. Although each of these analogies represents some key Hsp90 activities, none of them captures the entirety of Hsp90's complexity. Together, these roles enable Hsp90 to support both homeostasis and differentiation, both cellular stability and adaptability. At the 11th International Conference on the Hsp90 Chaperone Machine, the consensus was that to understand this major guardian of proteostasis, we need to study how the many facets of Hsp90's function influence each other. We hope that these analogies will help to conceptually integrate the many roles of Hsp90 in proteostasis and help the field develop the practical applications of Hsp90 modulators.
Collapse
Affiliation(s)
- Georgios Ioannis Karras
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston 77030, TX, USA; Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston 77030, TX, USA.
| | | | - Andrea N Kravats
- Department of Chemistry and Biochemistry, Miami University, Oxford 45056, OH, USA.
| |
Collapse
|
31
|
Karpagavalli M, Sindal MD, Arunachalam JP, Chidambaram S. miRNAs, piRNAs, and lncRNAs: A triad of non-coding RNAs regulating the neurovascular unit in diabetic retinopathy and their therapeutic potentials. Exp Eye Res 2025; 251:110236. [PMID: 39800284 DOI: 10.1016/j.exer.2025.110236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 12/04/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Diabetic Retinopathy (DR), a leading complication of diabetes mellitus, has long been considered as a microvascular disease of the retina. However, recent evidence suggests that DR is a neurovascular disease, characterized by the degeneration of retinal neural tissue and microvascular abnormalities encompassing ischemia, neovascularization, and blood-retinal barrier breakdown, ultimately leading to blindness. The intricate relationship between the retina and vascular cells constitutes a neurovascular unit, a multi-cellular framework of retinal neurons, glial cells, immune cells, and vascular cells, which facilitates neurovascular coupling, linking neuronal activity to blood flow. These interconnections between the neurovascular components get compromised due to hyperglycemia and are further associated with the progression of DR early on in the disease. As a result, therapeutic approaches are needed to avert the advancement of DR by acting at its initial stage to delay or prevent the pathogenesis. Non-coding RNAs (ncRNAs) such as microRNAs, piwi-interacting RNAs, and long non-coding RNAs regulate various cellular components in the neurovascular unit. These ncRNAs are key regulators of neurodegeneration, apoptosis, inflammation, and oxidative stress in DR. In this review, research related to alterations in the expression of ncRNAs and, correspondingly, their effect on the disintegration of the neurovascular coupling will be discussed briefly to understand the potential of ncRNAs as therapeutic targets for treating this debilitating disease.
Collapse
Affiliation(s)
| | | | - Jayamuruga Pandian Arunachalam
- Central Inter-Disciplinary Research Facility, Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India
| | - Subbulakshmi Chidambaram
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, 605 014, India.
| |
Collapse
|
32
|
Karatayli E, Sadiq SC, Schattenberg JM, Grabbe S, Biersack B, Kaps L. Curcumin and Its Derivatives in Hepatology: Therapeutic Potential and Advances in Nanoparticle Formulations. Cancers (Basel) 2025; 17:484. [PMID: 39941855 PMCID: PMC11816286 DOI: 10.3390/cancers17030484] [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: 12/20/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Curcumin, a plant-derived polyphenol, shows promise in hepatology for treating both malignant and non-malignant liver diseases and a subset of extrahepatic cancers. Curcumin has hepatoprotective, anti-inflammatory, antifibrotic, and antiproliferative properties, as is evident in preclinical and clinical studies. This highlights its potential as an adjunct to established cancer therapies, especially in the context of hepatocellular carcinoma and secondary liver malignancies. Curcumin also demonstrates potential in metabolic dysfunction-associated steatotic liver disease (MASLD), owing to its antifibrotic and lipid-lowering effects. However, its clinical use is limited, relating to its poor bioavailability and rapid metabolism. Nanotechnology, including liposomal and polymeric carriers, alongside synthetic curcumin derivatives, offers strategies to enhance the bioavailability and pharmacokinetic properties. We propose to revisit the use of curcumin in nanoparticle preparations in chronic liver disease and summarize current evidence in this review article.
Collapse
Affiliation(s)
- Ersin Karatayli
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (E.K.); (S.C.S.); (J.M.S.)
| | - Shifana C. Sadiq
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (E.K.); (S.C.S.); (J.M.S.)
| | - Jörn M. Schattenberg
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (E.K.); (S.C.S.); (J.M.S.)
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128 Mainz, Germany;
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, 95440 Bayreuth, Germany
| | - Leonard Kaps
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (E.K.); (S.C.S.); (J.M.S.)
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128 Mainz, Germany;
| |
Collapse
|
33
|
Wei J, Liu S, Bian Y, Li L, Qian B, Shen Z, Zhang Y, Abuduaini A, Dong F, Zhang X, Li J, Yu Y, Zhang W, Wang J, Zhai W, Song Q, Zheng Y, Pan W, Yu L, Zhan Q, Zhang N, Zheng J, Pan S, Yao C, Li H. Safety and efficacy of oral administrated cepharanthine in non-hospitalized, asymptomatic or mild COVID-19 patients: a Double-blind, randomized, placebo-controlled trial : Author detials. Sci Rep 2025; 15:3875. [PMID: 39890847 PMCID: PMC11785718 DOI: 10.1038/s41598-024-75891-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 10/09/2024] [Indexed: 02/03/2025] Open
Abstract
Cepharanthine (CEP) is a natural remedy that potently inhibits SARS-CoV-2 activity both in vitro and in vivo. To evaluate the efficacy and safety of CEP compared with placebo in adults with asymptomatic or mild coronavirus disease 2019 (COVID-19), we conducted a proof-of-concept, double-blind, randomized, placebo-controlled trial. Patients were randomized to receive 120 mg/day of CEP, 60 mg/day CEP or placebo for 5 days. Main outcome was the time from randomization to negative nasopharyngeal swab and safety. Among 262 randomized participants, 188 completed the trial among group of 120 mg/day CEP (n = 65), 60 mg/day CEP (n = 68) and placebo (n = 55). Neither 120 mg/day or 60 mg/day CEP shortened the time to negative significantly compared with placebo. However, 60 mg/day CEP showed a slight trend (difference=-0.77 days, hazard ratio (HR) = 1.40, 95% CI 0.97-2.01, p = 0.072). In analysis of participants with good medication compliance, 60 mg/day CEP significantly shortened the time to negative (difference=-0.87 days, HR = 1.56, 95% CI 1.03-2.37, p = 0.035). Adverse events were not different among the three groups, and no serious adverse events occurred. In conclusion, treatment of asymptomatic or mild Covid-19 with 120 mg/day or 60 mg/day did not shorten the time to negative significantly. However, 60 mg/day CEP showed a slight trend which needs future confirmatory trials to validate. (NCT05398705).
Collapse
Affiliation(s)
- Jianyi Wei
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Shupeng Liu
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Yuexiang Bian
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Lei Li
- Department of Otorhinolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Biyun Qian
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zixuan Shen
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Yan Zhang
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Adila Abuduaini
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Fuchen Dong
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Xin Zhang
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Jinhui Li
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Yongpei Yu
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, 100083, China
| | - Weituo Zhang
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Wang
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 160 Pujian Rd, Pudong, Shanghai, 200127, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qixiang Song
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zheng
- Department of Respiratory Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weihua Pan
- Department of Pediatric Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lanlan Yu
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, 100083, China
| | - Qimin Zhan
- Peking University - Yunnan Baiyao International Medical Research Center, Beijing, China
| | - Ning Zhang
- Peking University - Yunnan Baiyao International Medical Research Center, Beijing, China
| | - Junhua Zheng
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuming Pan
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China.
| | - Chen Yao
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, 100083, China.
| | - Hai Li
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China.
| |
Collapse
|
34
|
Alnagar AN, Motawea A, Zaghloul RA, Eldesoqui M, Abu Hashim II. A Novel Facile and Efficient Prophylaxis Avenue of Chitosan Oligosaccharide/PLGA Based Polydatin Loaded Nanoparticles Against Bleomycin-Induced Lung Inflammation in Experimental Rat Model. AAPS PharmSciTech 2025; 26:35. [PMID: 39820828 DOI: 10.1208/s12249-024-03022-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 12/05/2024] [Indexed: 01/19/2025] Open
Abstract
Lung inflammation is a hallmark of several respiratory diseases. Despite the great effectiveness of the synthetic antiinflammatory agents, they cause potential side effects. Polydatin (PD), a natural phytomedicine, has antioxidant and antiinflammatory effects. Its clinical applications are hindered due to poor aqueous solubility, low bioavailability, and rapid metabolism by first-pass effect. Herein, we report the development of a novel chitosan oligosaccharide-coated PD-loaded Poly dl-lactide-co-glycolide nanoparticles (COS-coated PD/PLGA NPs) against a bleomycin-induced pulmonary inflammation in a rat model. The NPs exhibited a small particle size of 188.57 ± 5.68 nm and a high zeta potential of + 18.13 ± 2.75 mV with spherical architecture and sustained release pattern of PD. In vivo studies in bleomycin-induced lung inflammation in a rat model revealed the superior prophylactic activity of COS-coated PD/PLGA NPs over the free drug (PD) as demonstrated by histopathological and immunohistochemical analyses, alongside biochemical assays evaluating oxidative stress biomarkers and inflammatory cytokine levels. Overall, the optimized COS-coated PD/PLGA NPs formulation offers a promising prophylactic platform against many respiratory diseases.
Collapse
Affiliation(s)
- Ahmed Nashaat Alnagar
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Dakahlia, Egypt
| | - Amira Motawea
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Dakahlia, Egypt.
| | - Randa A Zaghloul
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Dakahlia, Egypt
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, 13713, DiriyahRiyadh, Saudi Arabia
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Irhan Ibrahim Abu Hashim
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Dakahlia, Egypt
| |
Collapse
|
35
|
Eraky SM, Ramadan NM, Atif HM, Mahmoud AM, Abo El-Magd NF. The ameliorating effect of intermittent fasting on intestinal glucagon-like peptide 1 in rats fed a high-fat diet via the Farnesoid X receptor and the Melanocortin-4 receptor. Life Sci 2025; 361:123327. [PMID: 39716568 DOI: 10.1016/j.lfs.2024.123327] [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: 07/04/2024] [Revised: 11/08/2024] [Accepted: 12/19/2024] [Indexed: 12/25/2024]
Abstract
Obesity and its associated intestinal inflammatory responses represent a significant global challenge. (IF) is a dietary intervention demonstrating various health benefits, including weight loss, enhanced metabolic health, and increased longevity. However, its effect on the intestinal inflammation induced by high-fat diet (HFD) is still not fully comprehended. Thirty-four male Sprague-Dawley rats were randomized into three groups: Control (fed standard chow diet for 24 weeks); the HFD group (fed HFD for 24 weeks); and the HFD + IF group (fed HFD for 12 weeks, followed by an alternate day regimen of fasting and HFD for 12 weeks). The results revealed that IF significantly reduced body weight, food intake, and blood glucose levels compared to the HFD group. Furthermore, rats undergoing the intermittent fasting regimen exhibited a significant reduction in resting time, along with increased durations of grooming and exploration when compared to those on HFD. IF significantly reduced HFD-induced intestinal oxidative stress by lowering malondialdehyde levels and substantially increasing intestinal total antioxidant capacity, consistent with histopathological findings of gastric and intestinal tissues. The investigation of the underlying mechanisms revealed that IF significantly increased the intestinal expression of Farnesoid X receptor (FXR), glucagon-like peptide 1 (GLP-1), and melanocortin-4 receptors (MC4R), with a significant decrease in gastrointestinal peroxisome proliferator-activated receptor-γ (PPAR-γ) compared to the HFD group. The findings indicate that IF can mitigate HFD-induced intestinal inflammation via the FXR/GLP-1/MC4R/ PPAR-γ pathway. This highlights the need for further research to elucidate these mechanisms.
Collapse
Affiliation(s)
- Salma M Eraky
- Biochemistry Department, Faculty of Pharmacy, Mansoura University, Egypt
| | - Nehal M Ramadan
- Clinical Pharmacology Department, Faculty of Medicine, Mansoura University, Egypt
| | - Huda M Atif
- Medical Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Egypt
| | - Amr M Mahmoud
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El Aini, Cairo 11562, Egypt
| | - Nada F Abo El-Magd
- Biochemistry Department, Faculty of Pharmacy, Mansoura University, Egypt.
| |
Collapse
|
36
|
El-Zehery IM, El-Mesery M, El-Sherbiny M, El Gayar AM, Eisa NH. Carbenoxolone upregulates TRAIL\TRAILR2 expression and enhances the anti-neoplastic effect of doxorubicin in experimentally induced hepatocellular carcinoma in rats. Biochem Biophys Res Commun 2024; 741:150876. [PMID: 39579528 DOI: 10.1016/j.bbrc.2024.150876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/25/2024]
Abstract
AIMS This study investigates the in vivo anticancer activity of carbenoxolone (CBX) and its role in fighting hepatocellular carcinoma (HCC) progression and alleviating resistance against doxorubicin (DOX). Moreover, the molecular mechanism of action of CBX is explored. METHODS HCC was induced in Sprague Dawley rats via biweekly administration of thioacetamide (TAA) (200 mg/kg) intraperitoneally (i.p.) for 16 weeks after administering a single dose of diethylnitrosamine (DEN) (200 mg/kg, i.p.). A prophylactic model was established by treating rats with i.p. CBX (20 mg/kg/day) for 4 weeks starting on week 13 post-TAA injection. A therapeutic model was established by treating rats with CBX, DOX, or their combination for 7 weeks following 16 weeks of TAA administration. Serum Alpha-fetoprotein (AFP) and biochemical markers of hepatic functions were assessed. Histopathological examinations of hepatic tissues were performed. Immunohistochemical and qRT-PCR analyses were applied to assess the differential expressions of TRAIL/TRAILR2, Bcl-2, TGF-β1, and caspases 3, 8, and 9. RESULTS CBX markedly improved hepatic functions, reduced serum AFP levels, and alleviated TAA-induced hepatic histopathological alterations. CBX triggered apoptosis as evident by upregulating apoptotic markers: TRAIL/TRAILR2, caspases 3, 8, and 9, and downregulating the antiapoptotic protein Bcl-2. CBX downregulated TGF-β1. Interestingly, CBX/DOX combination mitigated hepatic damage and induced apoptosis in a way that surpassed DOX-only treatment. CONCLUSION The current study proposes that CBX is a promising anti-tumor compound, which can work effectively under prophylactic and therapeutic modes. Interestingly, CBX enhanced the anti-tumor effect of DOX. CBX exerted these effects via, in part, stimulating TRAIL-induced apoptosis along with attenuating fibrosis.
Collapse
MESH Headings
- Animals
- Doxorubicin
- Rats, Sprague-Dawley
- Male
- Carbenoxolone/pharmacology
- Rats
- Up-Regulation/drug effects
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/genetics
- Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
- Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/chemically induced
- Apoptosis/drug effects
- Antibiotics, Antineoplastic
- Drug Synergism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/chemically induced
- Liver Neoplasms/genetics
- Antineoplastic Agents/pharmacology
- TNF-Related Apoptosis-Inducing Ligand
Collapse
Affiliation(s)
- Iman M El-Zehery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Mohamed El-Mesery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt; Division of Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia; Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Amal M El Gayar
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Nada H Eisa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
37
|
Sharifi-Zahabi E, Abdollahzad H. Alpha Lipoic Acid Supplementation and Iron Homeostasis: A Comprehensive Systematic Review and Meta-Analysis of Randomized Controlled Clinical Trials. INT J VITAM NUTR RES 2024; 95:36623. [PMID: 40134249 DOI: 10.31083/ijvnr36623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 06/22/2024] [Accepted: 09/05/2024] [Indexed: 03/27/2025]
Abstract
BACKGROUND A growing body of evidence indicates the regulating effects of alpha-lipoic acid on iron metabolism. However, findings from clinical trials are equivocal. This systematic review and meta-analysis aimed to evaluate the quantitative effect of alpha lipoic acid (ALA) supplementation on iron metabolism parameters including serum iron, total iron binding capacity, hemoglobin, and ferritin. METHODOLOGY Online databases, including PubMed, Scopus, and Web of Science were searched, up to 29 May 2022, to obtain all relevant studies. RESULTS A total of 1901 publications were identified in the systematic search; of which, 10 studies with a total of 529 participants were included in this meta-analysis. Pooled analysis of the studies showed no statistically significant effects of ALA on ferritin (weighted mean difference (WMD) = -11.01 ng/mL; 95% CI: -40.07, 18.05 ng/mL; I2 = 0.0%, p = 0.670), serum iron (WMD = -0.47 μ/dL; 95% CI: -24.48, 23.54 μ/dL; I2 = 94.7%, p < 0.001), hemoglobin (WMD = 0.49 g/dL; 95% CI: -0.54, 1.52 g/dL; I2 = 95.7%, p < 0.001), and total iron binding capacity (TIBC) (WMD = 3.95 μ/dL; 95% CI: -21.3, 29.2 μ/dL; I2 = 53.1%, p = 0.094). In subgroup analysis, ALA significantly increased hemoglobin in patients with hematological disorders (WMD = 1.23 g/dL; 95% CI: 1.00, 1.45 g/dL; I2 = 96.6%, p < 0.001) and in studies with durations longer than 8 weeks (WMD = 1.03 g/dL; 95% CI: 0.82, 1.25 g/dL; I2 = 96.5%, p = 0.02). CONCLUSION ALA supplementation had no statistically significant effect on iron-related parameters. Subgroup analysis revealed a significant increasing effect of ALA on hemoglobin in patients with hematological disorders and in studies with durations >8 weeks.
Collapse
Affiliation(s)
- Elham Sharifi-Zahabi
- Student Research Committee, Kermanshah University of Medical Sciences, 6715847141 Kermanshah, Iran
| | - Hadi Abdollahzad
- School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, 6715847141 Kermanshah, Iran
| |
Collapse
|
38
|
García-Gurrola A, Martínez AL, Wall-Medrano A, Olivas-Aguirre FJ, Ochoa-Ruiz E, Escobar-Puentes AA. Phytochemistry, Anti-cancer, and Anti-diabetic Properties of Plant-Based Foods from Mexican Agrobiodiversity: A Review. Foods 2024; 13:4176. [PMID: 39767118 PMCID: PMC11675762 DOI: 10.3390/foods13244176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) and cancer are significant contributors to morbidity and mortality worldwide. Recent studies have increasingly highlighted the potential of phytochemicals found in plants and plant-based foods for preventing and treating these chronic diseases. Mexico's agrobiodiversity provides a valuable resource for phytochemistry. This review presents an examination of essential phytochemicals found in plants and foods within Mexican agrobiodiversity that have shown promising anti-cancer and anti-diabetic properties, including their roles as antioxidants, insulin sensitizers, and enzyme inhibitors. Notable compounds identified include flavonoids (such as quercetin and catechins), phenolic acids (chlorogenic, gallic, and caffeic acids), methylxanthines (like theobromine), xanthones (such as mangiferin), capsaicinoids (capsaicin), organosulfur compounds (like alliin), and various lipids (avocatins). Although these phytochemicals have shown promise in laboratory and animal studies, there is a significant scarcity of clinical trial data involving humans, underscoring an important area for future research.
Collapse
Affiliation(s)
- Adriana García-Gurrola
- Faculty of Medicine and Psychology, Autonomous University of Baja California, Tijuana 22427, Baja California, Mexico; (A.G.-G.); (A.L.M.); (E.O.-R.)
| | - Ana Laura Martínez
- Faculty of Medicine and Psychology, Autonomous University of Baja California, Tijuana 22427, Baja California, Mexico; (A.G.-G.); (A.L.M.); (E.O.-R.)
| | - Abraham Wall-Medrano
- Biomedical Sciences Institute, Autonomous University of Ciudad Juárez, Ciudad Juaez 32300, Chihuahua, Mexico; (A.W.-M.); (F.J.O.-A.)
| | - Francisco J. Olivas-Aguirre
- Biomedical Sciences Institute, Autonomous University of Ciudad Juárez, Ciudad Juaez 32300, Chihuahua, Mexico; (A.W.-M.); (F.J.O.-A.)
| | - Estefania Ochoa-Ruiz
- Faculty of Medicine and Psychology, Autonomous University of Baja California, Tijuana 22427, Baja California, Mexico; (A.G.-G.); (A.L.M.); (E.O.-R.)
| | - Alberto A. Escobar-Puentes
- Faculty of Medicine and Psychology, Autonomous University of Baja California, Tijuana 22427, Baja California, Mexico; (A.G.-G.); (A.L.M.); (E.O.-R.)
| |
Collapse
|
39
|
Morya AK, Ramesh PV, Nishant P, Kaur K, Gurnani B, Heda A, Salodia S. Diabetic retinopathy: A review on its pathophysiology and novel treatment modalities. World J Methodol 2024; 14:95881. [PMID: 39712561 PMCID: PMC11287547 DOI: 10.5662/wjm.v14.i4.95881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/28/2024] [Accepted: 07/10/2024] [Indexed: 07/26/2024] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic non-communicable disease with the ability to cause serious microvascular and macrovascular complications throughout the body, including in the eye. Diabetic retinopathy (DR), present in one-third of patients with diabetes, is a vision-threatening complication caused by uncontrolled diabetes, which greatly affects the retinal blood vessels and the light-sensitive inner retina, eventually leading to blindness. Several epidemiological studies elucidate that DR can vary by age of onset, duration, types of diabetes, and ethnicity. Recent studies show that the pathogenesis of diabetic retinopathy has spread its roots beyond merely being the result of hyperglycemia. The complexity of its etiopathology and diagnosis makes therapeutic intervention challenging. This review throws light on the pathological processes behind DR, the cascade of events that follow it, as well as the available and emerging treatment options.
Collapse
Affiliation(s)
- Arvind Kumar Morya
- Head of the Department, Department of Ophthalmology, All India Institute of Medical Sciences, Hyderabad 508126, Telangana, India
| | - Prasanna Venkatesh Ramesh
- Glaucoma Medical Officer, Department of Glaucoma and Research, Mahathma Eye Hospital Private Limited, Trichy 620017, Tamil Nadu, India
| | - Prateek Nishant
- Department of Ophthalmology, ESIC Medical College, Patna 801103, Bihar, India
| | - Kirandeep Kaur
- Department of Pediatric Ophthalmology and Strabismus, Gomabai Netralaya and Research Centre, Neemuch 458441, Madhya Pradesh, India
| | - Bharat Gurnani
- Cornea and Refractive Services, Gomabai Netralaya and Research Centre, Neemuch 458441, Madhya Pradesh, India
| | - Aarti Heda
- Department of Ophthalmology, National Institute of Ophthalmology, Pune 411000, Maharashtra, India
| | - Sarika Salodia
- Global Medical Safety, Lundbeck, Singapore 569933, Singapore, Singapore
| |
Collapse
|
40
|
Abdulal ZA, Altahhan MY, Qindil AF, Al-Juhani AM, Alatawi MA, Hassan HM, Al-Gayyar MM. Ferulic acid inhibits tumor proliferation and attenuates inflammation of hepatic tissues in experimentally induced HCC in rats. J Investig Med 2024; 72:900-910. [PMID: 39091053 DOI: 10.1177/10815589241270489] [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: 08/04/2024]
Abstract
Hepatocellular carcinoma (HCC) is a prevalent form of primary liver cancer with a 5-year survival rate of just 18%. Ferulic acid, a natural compound found in fruits and vegetables such as sweet corn, rice bran, and dong quai, is an encouraging drug known for its diverse positive effects on the body, including anti-inflammatory, anti-apoptotic, and neuroprotective properties. Our study aimed to investigate the potential antitumor effects of ferulic acid to inhibit tumor growth and inflammation of HCC in rats. HCC was induced in rats by administering thioacetamide. Additionally, some rats were given 50 mg/kg of ferulic acid three times a week for 16 weeks. Liver function was assessed by measuring serum alpha-fetoprotein (AFP) and examining hepatic tissue sections stained with hematoxylin/eosin or anti-hypoxia induced factor-1α (HIF-1α). The hepatic mRNA and protein levels of HIF-1α, nuclear factor κB (NFκB), tumor necrosis factor-α (TNF-α), mammalian target of rapamycin (mTOR), signal transducer and activator of transcription 3 (STAT3), cMyc, and cyclin D1 were examined. The results showed that ferulic acid increased the rats' survival rate by reducing serum AFP levels and suppressing hepatic nodules. Furthermore, ferulic acid ameliorated the appearance of vacuolated cytoplasm induced by HCC, reduced apoptotic nuclei, and necrotic nodules. Finally, ferulic acid decreased the expression of HIF-1α, NFκB, TNF-α, mTOR, STAT3, cMyc, and cyclin D1. In conclusion, ferulic acid is believed to possess antitumor properties by inhibiting HCC-induced hypoxia through the suppression of HIF-1α expression. Additionally, it helps in reducing the expression of mTOR, STAT3, cMyc, and cyclin D1, thereby slowing down tumor growth. Lastly, ferulic acid reduced hepatic tissue inflammation by downregulating NFκB and TNF-α.
Collapse
Affiliation(s)
| | | | | | | | | | - Hanan M Hassan
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City, Egypt
| | - Mohammed Mh Al-Gayyar
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
41
|
Hassan HM, Hamdan AM, Alattar A, Alshaman R, Bahattab O, Al-Gayyar MMH. Evaluating anticancer activity of emodin by enhancing antioxidant activities and affecting PKC/ADAMTS4 pathway in thioacetamide-induced hepatocellular carcinoma in rats. Redox Rep 2024; 29:2365590. [PMID: 38861483 PMCID: PMC11168332 DOI: 10.1080/13510002.2024.2365590] [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: 06/13/2024] Open
Abstract
Emodin is a naturally occurring anthraquinone derivative with a wide range of pharmacological activities, including neuroprotective and anti-inflammatory activities. We aim to assess the anticancer activity of emodin against hepatocellular carcinoma (HCC) in rat models using the proliferation, invasion, and angiogenesis biomarkers. After induction of HCC, assessment of the liver impairment and the histopathology of liver sections were investigated. Hepatic expression of both mRNA and protein of the oxidative stress biomarkers, HO-1, Nrf2; the mitogenic activation biomarkers, ERK5, PKCδ; the tissue destruction biomarker, ADAMTS4; the tissue homeostasis biomarker, aggregan; the cellular fibrinolytic biomarker, MMP3; and of the cellular angiogenesis biomarker, VEGF were measured. Emodin increased the survival percentage and reduced the number of hepatic nodules compared to the HCC group. Besides, emodin reduced the elevated expression of both mRNA and proteins of all PKC, ERK5, ADAMTS4, MMP3, and VEGF compared with the HCC group. On the other hand, emodin increased the expression of mRNA and proteins of Nrf2, HO-1, and aggrecan compared with the HCC group. Therefore, emodin is a promising anticancer agent against HCC preventing the cancer prognosis and infiltration. It works through many mechanisms of action, such as blocking oxidative stress, proliferation, invasion, and angiogenesis.
Collapse
Affiliation(s)
- Hanan M. Hassan
- Dept. of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Ahmed M. Hamdan
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdullah Alattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Reem Alshaman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Omar Bahattab
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohammed M. H. Al-Gayyar
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
42
|
Tülüce Y, Osmanoğlu D, Rağbetli MÇ, Altındağ F. Protective Action of Curcumin and Alpha-lipoic Acid, Against Experimental Ultraviolet-A/B Induced Dermal-injury in Rats. Cell Biochem Biophys 2024; 82:3535-3546. [PMID: 39060913 DOI: 10.1007/s12013-024-01442-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
The objective of this study was to examine the therapeutic efficacy of curcumin (CUR) and α-lipoic acid (ALA) in mitigating UV-A and UV-B-induced damage (UVAB) in rat dorsal skin. This was achieved through the utilisation of immunohistochemical (TUNEL), biochemical and stereological techniques. The rats in the UVAB, UVAB + CUR, and UVAB + ALA groups were subjected to UVAB irradiation for a period of two hours per day over the course of one month. The UVAB + CUR and UVAB + ALA groups were administered 100 mg/kg/day of curcumin and 100 mg/kg/day of α-lipoic acid via gavage 30 min prior to UVAB irradiation. The CUR group was administered 100 mg/kg/day of curcumin via gavage, while the ALA group received the same dose of α-lipoic acid. A significant change in the volume ratio of the dorsal skin epidermis and dermis was observed in the stereological findings of the rats in the UVAB group. These changes exhibited a favourable progression as a consequence of the CUR and ALA applications. In the UVAB group, TOS and OSI were significantly elevated as a consequence of the rise in oxidative stress. Conversely, the treatment groups demonstrated a notable reduction in TOS and OSI levels. The study also revealed a substantial increase in the number of apoptotic cells within the UVAB group. However, the treatment groups exhibited a significant decline in apoptotic cells. In conclusion, the findings suggest that CUR and ALA possess a protective effect against UVAB-induced skin damage.
Collapse
Affiliation(s)
- Yasin Tülüce
- Department of Medical Biology, Faculty of Medicine, Van Yüzüncü Yıl University, 65080, Van, Türkiye.
| | - Derya Osmanoğlu
- Department of Medical Histology and Embryology, Institute of Health Sciences, Van Yüzüncü Yıl University, 65080, Van, Türkiye
| | - Murat Çetin Rağbetli
- Department of Medical Histology and Embryology, Faculty of Medicine, Karamanoğlu Mehmetbey University, 70100, Karaman, Türkiye
| | - Fikret Altındağ
- Department of Medical Histology and Embryology, Faculty of Medicine, Van Yüzüncü Yıl University, 65080, Van, Türkiye
| |
Collapse
|
43
|
Pu D, Wang Z, Zheng J, Li P, Wei X, Li D, Gao L, Zhou L, Wang Y. Effects of Ammonia Stress on Liver Tissue Structure, Enzyme Activities, and Metabolome of Juvenile Largemouth Bass Micropterus salmoides. Metabolites 2024; 14:649. [PMID: 39728430 DOI: 10.3390/metabo14120649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024] Open
Abstract
Background: Ammonia, a ubiquitous contaminant in aquatic ecosystems, poses multifaceted threats to fish species at elevated concentrations. Methods: In order to investigate the toxic effects of chronic ammonia stress on the liver of juvenile Micropterus salmoides, the present experiment was conducted to investigate the differences in changes in liver tissue structure, enzyme activities, and metabolomes after 28 days of ammonia exposure (0, 4, 8, and 16 mg/L). Results: The findings revealed that ammonia exposure induced significant oxidative stress in the liver, manifesting in decreased activities of antioxidant enzymes SOD and GSH-Px, elevated levels of GSH, GST, and MDA, and heightened activities of immune enzymes LZM, ALP, and ACP. An increase in ammonia concentration exacerbated liver tissue damage. Metabolome analysis further unveiled perturbations in liver metabolites of Micropterus salmoides exposed to ammonia, with Ala-His emerging as a potentially pivotal functional substance under chronic stress. Specifically, the 4 mg/L group responded to ammonia toxicity by augmenting GSH and L-Carnosine levels, the 8 mg/L group detoxified via upregulation of L-Glutamine, and the 16 mg/L group mitigated toxicity through the urea synthesis pathway. Conclusions: This research offers preliminary insights into the toxicological responses of Micropterus salmoides under chronic ammonia stress. It is suggested that the duration of ammonia concentration exceeding 4 mg/L in high-density aquaculture should not exceed 7 days.
Collapse
Affiliation(s)
- Decheng Pu
- Key Laboratory of Smart Agricultural Technology in the Southwest Mountains, Ministry of Agriculture and Rural Affairs (Co-Construction by Ministry and Province), Chongqing Academy of Agricultural Sciences, Chongqing 400715, China
- College of Fisheries, Southwest University, Chongqing 400715, China
| | - Zhengxi Wang
- Key Laboratory of Smart Agricultural Technology in the Southwest Mountains, Ministry of Agriculture and Rural Affairs (Co-Construction by Ministry and Province), Chongqing Academy of Agricultural Sciences, Chongqing 400715, China
| | - Jishu Zheng
- Key Laboratory of Smart Agricultural Technology in the Southwest Mountains, Ministry of Agriculture and Rural Affairs (Co-Construction by Ministry and Province), Chongqing Academy of Agricultural Sciences, Chongqing 400715, China
| | - Peiyuan Li
- Key Laboratory of Smart Agricultural Technology in the Southwest Mountains, Ministry of Agriculture and Rural Affairs (Co-Construction by Ministry and Province), Chongqing Academy of Agricultural Sciences, Chongqing 400715, China
| | - Xiuli Wei
- Key Laboratory of Smart Agricultural Technology in the Southwest Mountains, Ministry of Agriculture and Rural Affairs (Co-Construction by Ministry and Province), Chongqing Academy of Agricultural Sciences, Chongqing 400715, China
| | - Dongsheng Li
- Key Laboratory of Smart Agricultural Technology in the Southwest Mountains, Ministry of Agriculture and Rural Affairs (Co-Construction by Ministry and Province), Chongqing Academy of Agricultural Sciences, Chongqing 400715, China
| | - Lihong Gao
- Key Laboratory of Smart Agricultural Technology in the Southwest Mountains, Ministry of Agriculture and Rural Affairs (Co-Construction by Ministry and Province), Chongqing Academy of Agricultural Sciences, Chongqing 400715, China
| | - Lin Zhou
- Key Laboratory of Smart Agricultural Technology in the Southwest Mountains, Ministry of Agriculture and Rural Affairs (Co-Construction by Ministry and Province), Chongqing Academy of Agricultural Sciences, Chongqing 400715, China
| | - Yu Wang
- Key Laboratory of Smart Agricultural Technology in the Southwest Mountains, Ministry of Agriculture and Rural Affairs (Co-Construction by Ministry and Province), Chongqing Academy of Agricultural Sciences, Chongqing 400715, China
| |
Collapse
|
44
|
Karger G, Willker JE, Harders AR, Watermann P, Dringen R. ATP Restoration by ATP-Deprived Cultured Primary Astrocytes. Neurochem Res 2024; 50:13. [PMID: 39549173 PMCID: PMC11569012 DOI: 10.1007/s11064-024-04276-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 11/18/2024]
Abstract
A high cellular concentration of adenosine triphosphate (ATP) is essential to fuel many important functions of brain astrocytes. Although cellular ATP depletion has frequently been reported for astrocytes, little is known on the metabolic pathways that contribute to ATP restoration by ATP-depleted astrocytes. Incubation of cultured primary rat astrocytes in glucose-free buffer for 60 min with the mitochondrial uncoupler BAM15 lowered the cellular ATP content by around 70%, the total amount of adenosine phosphates by around 50% and the adenylate energy charge (AEC) from 0.9 to 0.6. Testing for ATP restoration after removal of the uncoupler revealed that the presence of glucose as exclusive substrate allowed the cells to restore within 6 h around 80% of the initial ATP content, while coapplication of adenosine plus glucose enabled the cells to fully restore their initial ATP content within 60 min. A rapid but incomplete and transient ATP restoration was found for astrocytes that had been exposed to adenosine alone. This restoration was completely prevented by application of the pyruvate uptake inhibitor UK5099, the respiratory chain inhibitor antimycin A or by the continuous presence of BAM15. However, the presence of these compounds strongly accelerated the release of lactate from the cells, suggesting that the ribose moiety of adenosine can serve as substrate to fuel some ATP restoration via mitochondrial metabolism. Finally, the adenosine-accelerated ATP restoration in glucose-fed astrocytes was inhibited by the presence of the adenosine kinase inhibitor ABT-702. These data demonstrate that astrocytes require for a rapid and complete ATP restoration the presence of both glucose as substrate and adenosine as AMP precursor.
Collapse
Affiliation(s)
- Gabriele Karger
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Johanna Elisabeth Willker
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Antonia Regina Harders
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Patrick Watermann
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany.
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany.
| |
Collapse
|
45
|
Xiao J, Xu Z. Roles of noncoding RNAs in diabetic retinopathy: Mechanisms and therapeutic implications. Life Sci 2024; 357:123092. [PMID: 39368772 DOI: 10.1016/j.lfs.2024.123092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/20/2024] [Accepted: 09/28/2024] [Indexed: 10/07/2024]
Abstract
Diabetic retinopathy (DR) is a microvascular complication of diabetes that leads to vision loss. The striking features of DR are hard exudate, cotton-wool spots, hemorrhage, and neovascularization. The dysregulated retinal cells, encompassing microvascular endothelial cells, pericytes, Müller cells, and adjacent retinal pigment epithelial cells, are involved in the pathological processes of DR. According to recent research, oxidative stress, inflammation, ferroptosis, pyroptosis, apoptosis, and angiogenesis contribute to DR. Recent advancements have highlighted that noncoding RNAs could regulate diverse targets in pathological processes that contribute to DR. Noncoding RNAs, including long noncoding RNAs, microRNAs (miRNA), and circular RNAs, are dysregulated in DR, and interact with miRNA, mRNA, or proteins to control the pathological processes of DR. Hence, modulation of noncoding RNAs may have therapeutic effects on DR. Small extracellular vesicles may be valuable tools for transferring noncoding RNAs and regulating the genes involved in progression of DR. However, the roles of noncoding RNA in developing DR are not fully understood; it is critical to summarize the mechanisms for noncoding RNA regulation of pathological processes and pathways related to DR. This review provides a fundamental understanding of the relationship between noncoding RNAs and DR, exploring the mechanism of how noncoding RNA modulates different signaling pathways, and pave the way for finding potential therapeutic strategies for DR.
Collapse
Affiliation(s)
- Jing Xiao
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhuping Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
46
|
Su Q, Sun H, Mei L, Yan Y, Ji H, Chang L, Wang L. Ribosomal proteins in hepatocellular carcinoma: mysterious but promising. Cell Biosci 2024; 14:133. [PMID: 39487553 PMCID: PMC11529329 DOI: 10.1186/s13578-024-01316-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
Ribosomal proteins (RPs) are essential components of ribosomes, playing a role not only in ribosome biosynthesis, but also in various extra-ribosomal functions, some of which are implicated in the development of different types of tumors. As universally acknowledged, hepatocellular carcinoma (HCC) has been garnering global attention due to its complex pathogenesis and challenging treatments. In this review, we analyze the biological characteristics of RPs and emphasize their essential roles in HCC. In addition to regulating related signaling pathways such as the p53 pathway, RPs also act in proliferation and metastasis by influencing cell cycle, apoptosis, angiogenesis, and epithelial-to-mesenchymal transition in HCC. RPs are expected to unfold new possibilities for precise diagnosis and individualized treatment of HCC.
Collapse
Affiliation(s)
- Qian Su
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Huizhen Sun
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Ling Mei
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Ying Yan
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Huimin Ji
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Le Chang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China.
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China.
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China.
| | - Lunan Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China.
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China.
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China.
| |
Collapse
|
47
|
Baig SI, Naseer M, Munir AR, Ali Y, Razzaq MA. Immunoinformatic-based drug design utilizing hesperetin to target CISD2 activation for liver aging in humans. Biogerontology 2024; 25:1189-1213. [PMID: 39196437 DOI: 10.1007/s10522-024-10130-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024]
Abstract
The CISD protein family, consisting of CISD1, CISD2, and CISD3, encodes proteins that feature CDGSH iron-sulfur domains crucial for cellular functions and share a common 2Fe-2S domain. CISD2, which is pivotal in cells, regulates intracellular calcium levels, maintains the endoplasmic reticulum and mitochondrial function, and is associated with longevity and overall health, with exercise stimulating CISD2 production. However, CISD2 expression decreases with age, impacting age-related processes. According to in silico docking, HST is a CISD2 activator that affects metabolic dysfunction and age-related illnesses by affecting metabolic pathways. This study investigated the ability of CISD2 and HST to reduce age-related ailments, with a particular emphasis on liver aging. CISD2 deficiency has a major effect on the function of cells, as it undermines the integrity of the ER, mitochondria, and calcium homeostasis. It also increases susceptibility to oxidative stress and metabolic dysregulation, which is linked to Wolfram syndrome and exacerbates age-related illnesses and metabolic disorders. By shielding cells from stress, CISD2 extends the life of cells and maintains liver health as people age. Its protective effecfts on the liver during aging are further enhanced by its control of translation factors such as Nrf2 and IL-6. This work paves the way for future investigations and clinical applications by examining the structural and functional properties of CISD2 and the interaction between CISD2 and HST. This highlights the therapeutic potential of these findings in promoting healthy livers in humans and battling age-related illnesses.
Collapse
Affiliation(s)
- Saad Ilyas Baig
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan.
| | - Maria Naseer
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan
| | - Abdur-Rehman Munir
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan
| | - Yasir Ali
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan
| | | |
Collapse
|
48
|
El-Masry TA, El-Nagar MMF, Oriquat GA, Alotaibi BS, Saad HM, El Zahaby EI, Ibrahim HA. Therapeutic efficiency of Tamoxifen/Orlistat nanocrystals against solid ehrlich carcinoma via targeting TXNIP/HIF1-α/MMP-9/P27 and BAX/Bcl2/P53 signaling pathways. Biomed Pharmacother 2024; 180:117429. [PMID: 39293373 DOI: 10.1016/j.biopha.2024.117429] [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: 06/27/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Orlistat (Orli) is an anti-obesity medication that has been approved by the US Food and Drug Administration. It has relatively limited oral bioavailability with promising inhibitory effects on cell proliferation as well as reducing the growth of tumors. AIMS This investigation was done to evaluate the potential protective effect of Tamoxifen/Orlistat nanocrystals alone or in combination against Solid Ehrlich Carcinoma (SEC) and to clarify the possible underlying influences. MATERIALS AND METHODS The liquid antisolvent precipitation technique (bottom-up technology) was utilized to manufacture Orlistat Nanocrystals. To explore potential causes for the anti-tumor action, female Swiss Albino mice bearing SEC were randomly assigned into five equal groups (n = 6). Group 1: Tumor control group, group 2: Tam group: tamoxifen (0.01 g/kg, IP), group 3: Free-Orli group: orlistat (0.24 g/kg, IP), group 4: Nano-Orli: orlistat nanocrystals (0.24 g/kg, IP), group 5: Tam-Nano-Orli: Both doses of Tam and Nano-Orli. All treatments were administered for 16 days. KEY FINDINGS The untreated mice showed development in the tumor volume and weight. As well as histopathology results from these mice revealed many tumor large cells as well as solid sheets of malignant cells. Also, untreated mice showed raised VEGF and TGF-1beta content. Moreover, results of gene expression in the SEC-bearing mice noted upregulation in HIF-1α, MMP-9, Bcl-2, and P27 gene expression and downregulation of TXNIP, BAX, and P53 gene expression. On the other hand, administrated TAM, Free-Orli, Nano-Orli, and a combination of Tam-Nano-Orli distinctly suppressed the tumor effects on estimated parameters with special reference to Tam-Nano-Orli. SIGNIFICANCE The developed Tamoxifen/Orlistat nanocrystals combination could be considered a promising approach to augment antitumor effects.
Collapse
Affiliation(s)
- Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Maysa M F El-Nagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Ghaleb Ali Oriquat
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan.
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Cairo 51511, Egypt.
| | - Enas I El Zahaby
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 35712, Egypt.
| | - Hanaa A Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
49
|
Elazab ST, Eldin REAG. α-Bisabolol and royal jelly differentially mitigate thioacetamide-induced hepatic fibrosis in rats associated with the inhibition of TGF-β1/FAK/α-SMA signaling. Food Chem Toxicol 2024; 193:115069. [PMID: 39442747 DOI: 10.1016/j.fct.2024.115069] [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: 08/03/2024] [Revised: 10/13/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Hepatic fibrosis is a global health burden that accounts for high mortality. No definitive therapy to suppress the fibrosis so far. Thus, looking for an effective remedy to address the unmet medical need is crucial. We aimed to scrutinize the efficacy of royal jelly (RJ) and/or α-Bisabolol (BISA) in the regression of fibrosis provoked by thioacetamide (TAA), focusing on their action on redox status, NF-κBp65, apoptosis, and TGF-β1/FAK/α-SMA pathway. TAA was injected intraperitoneally twice weekly to trigger hepatic fibrosis. Rats were gavaged with RJ (100 mg/kg) and/or BISA (50 mg/kg) daily for 8 weeks. The findings elucidated that RJ and/or BISA alleviated TAA-provoked fibrosis mirrored by the improvement of hepatotoxicity serum indices, abolishing oxidative stress, and repair the morphological alterations. Additionally, RJ and BISA suppressed the hepatic inflammation induced by TAA through downregulating NF-κBp65 expression, reducing TNF-α and IL-6 concentrations, and elevating IL-10 level. Their anti-fibrotic effect was emphasized from the decline in FAK, Smad3, COL-III, hydroxyproline levels, and TGF-β1, α-SMA immunoexpression. BISA displayed better ameliorative action than RJ. Conclusively, RJ and/or BISA possess a hepatoprotective activity against TAA-mediated fibrosis by enhancing antioxidant defense, inhibiting NF-κBp65, and modulating TGF-β1/FAK/α-SMA signaling. RJ and BISA might be prospective candidates to combat hepatic fibrosis.
Collapse
Affiliation(s)
- Sara T Elazab
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | | |
Collapse
|
50
|
Jagdale AD, Angal MM, Patil RS, Tupe RS. Exploring the glycation association with dyslipidaemia: Novel approach for diabetic nephropathy. Biochem Pharmacol 2024; 229:116513. [PMID: 39218042 DOI: 10.1016/j.bcp.2024.116513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/14/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
The transcription factor known as sterol regulatory element-binding protein (SREBP) and the glycation pathways, specifically the formation of Advanced Glycation End Products (AGEs), have a significant and deleterious impact on the kidney. They alter renal lipid metabolism and promote glomerulosclerosis, mesangial cell expansion, tubulointerstitial fibrosis, and inflammation, leading to diabetic nephropathy (DN) progression. Although several pieces of scientific evidence are reported for potential causes of glycation and lipotoxicity in DN, the underlying mechanism of renal lipid accumulation still needs to be fully understood. We provide a rationalized view on how AGEs exert multiple effects that cause SREBP activation and inflammation, contributing to DN through Receptor for AGEs (RAGE) signaling, AGE-R1-dependent downregulation of Sirtuin 1 (SIRT-1), and increased SREBP Cleavage Activating Protein (SCAP) glycosylation. This review emphasizes the association between glycation and the SREBP pathway and how it affects the onset of DN associated with obesity. Finally, we discuss the correlation of glycation and the SREBP pathway with insulin resistance (IR), oxidative stress, endoplasmic reticulum stress, inflammation, and existing and emerging therapeutic approaches toward better controlling obesity-related DN.
Collapse
Affiliation(s)
- Ashwini D Jagdale
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| | - Mukul M Angal
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| | - Rahul S Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India.
| |
Collapse
|