1
|
Li C, Wang Y, Liang J, Haenen GRMM, Chen Y, Li Z, Zhang M, Dubois LJ. Exploring the Anticancer Potential of MonoHER (7-Mono-O-(β-Hydroxyethyl)-Rutoside): Mitochondrial-Dependent Apoptosis in HepG2 Cells. Curr Issues Mol Biol 2025; 47:36. [PMID: 39852151 PMCID: PMC11763755 DOI: 10.3390/cimb47010036] [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: 12/11/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/AIM Flavonoids are a group of polyphenols, abundantly present in our diet. Although, based on their chemoprotective effects, intake of flavonoids is associated with a high anticancer potential as evidenced in in vitro and in vivo models, the molecular mechanism is still elusive. This study explores the antiproliferative and cytotoxic effects of the semi-synthetic flavonoid MonoHER (7-mono-O-(β-hydroxyethyl)-rutoside) in vitro on cancer cells. MATERIALS AND METHODS HepG2 liver, MCF7 breast, and H1299 lung cancer cells were grown under ambient conditions with or without MonoHER exposure. CCK8 assay was used to assess cell viability. Apoptosis, JC-1, and mitochondrial mass were determined using flow cytometry and confocal analysis. The effects of monoHER on apoptosis proteins were detected by confocal microscopy analysis and Western blot. RESULTS It was found that MonoHER can reduce HepG2 cells' and MCF7 cells' viability, but not H1299 cells', and induced apoptosis only in HepG2 cells. MonoHER has the potential to enhance the expression of caspase-9 and caspase-3, to damage mitochondria, and to provoke the release of cytochrome C from the mitochondria. CONCLUSION MonoHER can inhibit cell growth and induce apoptosis especially in HepG2 human liver cancer cells by triggering the mitochondrial signal transduction pathway, leading to the release of cytochrome C in the cytoplasm and the subsequent activation of caspase-9 and caspase-3. Future research should further explore MonoHER's mechanism of action, efficacy, and potential for clinical translation.
Collapse
Affiliation(s)
- Chujie Li
- Department of Pharmacology and Personalized Medicine, Research Institute for Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200MD Maastricht, The Netherlands; (C.L.); (Y.W.); (G.R.M.M.H.)
- The M-Lab, Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6200MD Maastricht, The Netherlands
| | - Yue Wang
- Department of Pharmacology and Personalized Medicine, Research Institute for Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200MD Maastricht, The Netherlands; (C.L.); (Y.W.); (G.R.M.M.H.)
| | - Jian Liang
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China;
| | - Guido R. M. M. Haenen
- Department of Pharmacology and Personalized Medicine, Research Institute for Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200MD Maastricht, The Netherlands; (C.L.); (Y.W.); (G.R.M.M.H.)
| | - Yonger Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China;
| | - Zhengwen Li
- School of Pharmacy, Chengdu University, 2025 Chengluo Avenue, Chengdu 610106, China;
| | - Ming Zhang
- Hainan University-HSF/LWL Collaborative Innovation Laboratory, College of Food Sciences & Engineering, Hainan University, 58 People Road, Haikou 570228, China
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6200MD Maastricht, The Netherlands
| |
Collapse
|
2
|
Mancuso G, Violi F, Nocella C. Food contamination and cardiovascular disease: a narrative review. Intern Emerg Med 2024; 19:1693-1703. [PMID: 38743129 PMCID: PMC11405437 DOI: 10.1007/s11739-024-03610-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 04/09/2024] [Indexed: 05/16/2024]
Abstract
Cardiovascular disease is a significant cause of morbidity and mortality among non-communicable diseases worldwide. Evidence shows that a healthy dietary pattern positively influences many risk factors of cardiometabolic health, stroke, and heart disease, supported by the effectiveness of healthy diet and lifestyles for the prevention of CVD. High quality and safety of foods are prerequisites to ensuring food security and beneficial effects. Contaminants can be present in foods mainly because of contamination from environmental sources (water, air, or soil pollution), or artificially introduced by the human. Moreover, the cross-contamination or formation during food processing, food packaging, presence or contamination by natural toxins, or use of unapproved food additives and adulterants. Numerous studies reported the association between food contaminants and cardiovascular risk by demonstrating that (1) the cross-contamination or artificial sweeteners, additives, and adulterants in food processing can be the cause of the risk for major adverse cardiovascular events and (2) environmental factors, such as heavy metals and chemical products can be also significant contributors to food contamination with a negative impact on cardiovascular systems. Furthermore, oxidative stress can be a common mechanism that mediates food contamination-associated CVDs as substantiated by studies showing impaired oxidative stress biomarkers after exposure to food contaminants.This narrative review summarizes the data suggesting how food contaminants may elicit artery injury and proposing oxidative stress as a mediator of cardiovascular damage.
Collapse
Affiliation(s)
- Gerardo Mancuso
- Internal Medicine Unit, Department of Medicine and Medical Specialties, Lamezia Terme Hospital, 88046, Lamezia Terme, Italy
| | - Francesco Violi
- Department of Clinical Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Cristina Nocella
- Department of Clinical Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161, Rome, Italy.
| |
Collapse
|
3
|
Zhou Y, Jin W, Wu Q, Zhou Q. Acrolein: formation, health hazards and its controlling by dietary polyphenols. Crit Rev Food Sci Nutr 2024; 64:9604-9617. [PMID: 37203991 DOI: 10.1080/10408398.2023.2214625] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Acrolein, a highly reactive toxic aldehyde, is a common dietary and environmental contaminant which can also be generated endogenously. Exposure to acrolein has been positively associated with some pathological conditions, such as atherosclerosis, diabetes mellitus, stroke, and Alzheimer's disease. At the cellular level, acrolein induces various harmful effects, particularly protein adduction and oxidative damages. Polyphenols are a group of secondary plant metabolites ubiquitously presented in fruits, vegetables, and herbs. Recent evidence has gradually solidified the protective role of polyphenols by working as acrolein scavengers and regulator of acrolein toxicities. This was largely attributed to the ability of polyphenols as antioxidants and sacrificial nucleophiles in trapping acrolein. This review discussed the exposure and toxicity of acrolein, summarized the known and anticipated contribution of polyphenols in ameliorating acrolein contamination and its health hazards.
Collapse
Affiliation(s)
- Yue Zhou
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Advanced Study, Shenzhen University, Shenzhen, China
| | - Wendy Jin
- Rutgers Core Facility for Natural Products and Bioanalysis, New Use Agriculture and Natural Plant Products Program (NUANP), Department of Plant Biology, Rutgers University, New Brunswick, NJ, USA
| | - Qingli Wu
- Rutgers Core Facility for Natural Products and Bioanalysis, New Use Agriculture and Natural Plant Products Program (NUANP), Department of Plant Biology, Rutgers University, New Brunswick, NJ, USA
| | - Qian Zhou
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Advanced Study, Shenzhen University, Shenzhen, China
| |
Collapse
|
4
|
Ji Y, Morel Y, Tran AQ, Lipinski MM, Sarkar C, Jones JW. Development and evaluation of a liquid chromatography-tandem mass spectrometry method for simultaneous measurement of toxic aldehydes from brain tissue. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1242:124208. [PMID: 38880056 PMCID: PMC11227393 DOI: 10.1016/j.jchromb.2024.124208] [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: 04/09/2024] [Revised: 05/24/2024] [Accepted: 06/11/2024] [Indexed: 06/18/2024]
Abstract
Reactive aldehydes are a class of electrophilic low molecular weight compounds that play an essential role in physiological function and lipid peroxidation. These molecules are implicated in many diseases, especially cardiovascular and neurodegenerative diseases, and are potential endogenous markers of lipid peroxidation. However, there are limited options to accurately quantify multiple reactive aldehydes in brain tissue. This study developed and validated a 3-nitrophenylhydrazine derivatization-based LC-MS/MS method to quantify four reactive aldehydes: malondialdehyde, acrolein, 4-hydroxy-2-hexenal and 4-hydroxy-2-nonenal. Method development involved comparing the sensitivity of detection between widely used derivatization reagents: 2,4-dinitrophenylhydrazine and 3-nitrophenylhydrazine. Our data showed that 3-nitrophenylhydrazine resulted in greater sensitivity. Additional method development included evaluation of hydrolysis sample pretreatment, selection of protein precipitation reagent, and optimization of derivatization conditions. The optimized conditions included no hydrolysis and use of 20 % trichloroacetic acid as the protein precipitation reagent. The optimized derivatization condition was 25 mM 3-nitrophenylhydrazine reacted at 20 °C for 30 min. The chromatographic conditions were optimized to reduce matrix effects, ion suppression, and efficient analysis time (<7-minute analytical run). The four aldehyde species were accurately quantified in brain tissue using stable-labeled internal standards. Application of this assay to a traumatic brain injury mouse model revealed significant accumulation of acrolein, 4-hydroxy-2-hexenal, and 4-hydroxy-2-nonenal at 28 days post injury. Overall, a validated method was developed to rapidly quantify the most prominent reactive aldehydes associated with lipid peroxidation during injury progression following acute brain trauma.
Collapse
Affiliation(s)
- Yuanyuan Ji
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Yulemni Morel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Anh Q Tran
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Marta M Lipinski
- Department of Anesthesiology, Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chinmoy Sarkar
- Department of Anesthesiology, Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jace W Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.
| |
Collapse
|
5
|
Fujiwara G, Okada Y, Shiomi N, Sakakibara T, Yamaki T, Hashimoto N. Derivation of Coagulation Phenotypes and the Association with Prognosis in Traumatic Brain Injury: A Cluster Analysis of Nationwide Multicenter Study. Neurocrit Care 2024; 40:292-302. [PMID: 36977962 DOI: 10.1007/s12028-023-01712-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 03/01/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND The pathogenesis and pathophysiology of traumatic coagulopathy during traumatic brain injury is not well understood, and the appropriate treatment strategy for this condition has not been established. This study aimed to evaluate the coagulation phenotypes and their effect on prognosis in patients with isolated traumatic brain injury. METHODS In this multicenter cohort study, we retrospectively analyzed data from the Japan Neurotrauma Data Bank. Adults with isolated traumatic brain injury (head abbreviated injury scale > 2; abbreviated injury scale of any other trauma < 3) who were registered in the Japan Neurotrauma Data Bank were included in this study. The primary outcome was the association of coagulation phenotypes with in-hospital mortality. Coagulation phenotypes were derived using k-means clustering with coagulation markers, including prothrombin time international normalized ratio (PT-INR), activated partial thromboplastin time (APTT), fibrinogen (FBG), and D-dimer (DD) on arrival at the hospital. Multivariable logistic regression analyses were conducted to calculate the adjusted odds ratios of coagulation phenotypes with their 95% confidence intervals (CIs) for in-hospital mortality. RESULTS In total, 556 patients were enrolled and five coagulation phenotypes were identified. The median (interquartile range) score for the Glasgow Coma Scale was 6 (4-9). Cluster A (n = 129) had the closest to normal coagulation values; cluster B (n = 323) had a mild high DD phenotype; cluster C (n = 30) had a prolonged PT-INR phenotype with a higher frequency of antithrombotic medication in elderly patients than in younger patients; cluster D (n = 45) had a low amount of FBG, high DD, and prolonged APTT phenotype with a high incidence of skull fracture; and cluster E (n = 29) had a low amount of FBG and extremely high DD phenotype with high energy trauma and a high incidence of skull fracture. In the multivariable logistic regression analysis, the association of clusters B, C, D, and E with in-hospital mortality yielded the corresponding adjusted odds ratios of 2.17 (95% CI 1.22-3.86), 2.61 (95% CI 1.01-6.72), 10.0 (95% CI 4.00-25.2), and 24.1 (95% CI 7.12-81.3), respectively, relative to cluster A. CONCLUSIONS This multicenter, observational study identified five different coagulation phenotypes of traumatic brain injury and showed associations of these phenotypes with in-hospital mortality.
Collapse
Affiliation(s)
- Gaku Fujiwara
- Department of Neurosurgery, Saiseikai Shiga Hospital, Imperial Gift Foundation Inc, 2-4-1, Ohashi, Ritto, Shiga, Japan.
| | - Yohei Okada
- Department of Preventive Services, School of Public Health, Kyoto University, Kyoto, Japan
| | - Naoto Shiomi
- Department of Critical and Intensive Care Medicine, Shiga University of Medical Science, Ritto, Shiga, Japan
| | | | - Tarumi Yamaki
- Department of Neurosurgery, Kyoto Kujo Hospital, Kyoto, Japan
| | - Naoya Hashimoto
- Department of Neurosurgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
6
|
Kempuraj D, Mohan RR. Autophagy in Extracellular Matrix and Wound Healing Modulation in the Cornea. Biomedicines 2022; 10:biomedicines10020339. [PMID: 35203548 PMCID: PMC8961790 DOI: 10.3390/biomedicines10020339] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Autophagy is a robust cellular mechanism for disposing of harmful molecules or recycling them to cells, which also regulates physiopathological processes in cornea. Dysregulated autophagy causes inefficient clearance of unwanted proteins and cellular debris, mitochondrial disorganization, defective inflammation, organ dysfunctions, cell death, and diseases. The cornea accounts for two-thirds of the refraction of light that occurs in the eyes, but is prone to trauma/injury and infection. The extracellular matrix (ECM) is a noncellular dynamic macromolecular network in corneal tissues comprised of collagens, proteoglycans, elastin, fibronectin, laminins, hyaluronan, and glycoproteins. The ECM undergoes remodeling by matrix-degrading enzymes and maintains corneal transparency. Autophagy plays an important role in the ECM and wound healing maintenance. Delayed/dysregulated autophagy impacts the ECM and wound healing, and can lead to corneal dysfunction. Stromal wound healing involves responses from the corneal epithelium, basement membrane, keratocytes, the ECM, and many cytokines and chemokines, including transforming growth factor beta-1 and platelet-derived growth factor. Mild corneal injuries self-repair, but greater injuries lead to corneal haze/scars/fibrosis and vision loss due to disruptions in the ECM, autophagy, and normal wound healing processes. Presently, the precise role of autophagy and ECM remodeling in corneal wound healing is elusive. This review discusses recent trends in autophagy and ECM modulation in the context of corneal wound healing and homeostasis.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65212, USA;
- One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Rajiv R. Mohan
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65212, USA;
- One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- Correspondence:
| |
Collapse
|
7
|
Qiu Z, He J, Shao G, Hu J, Li X, Zhou H, Li M, Yang B. Obacunone Retards Renal Cyst Development in Autosomal Dominant Polycystic Kidney Disease by Activating NRF2. Antioxidants (Basel) 2021; 11:antiox11010038. [PMID: 35052542 PMCID: PMC8773305 DOI: 10.3390/antiox11010038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/19/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common inherited disease characterized by progressive enlargement of fluid-filled cysts derived from renal tubular epithelial cells, which has become the fourth leading cause of end-stage renal diseases. Currently, treatment options for ADPKD remain limited. The purpose of this study was to discover an effective therapeutic drug for ADPKD. With virtual screening, Madin-Darby canine kidney (MDCK) cyst model, embryonic kidney cyst model and kidney-specific Pkd1 knockout mouse (PKD) model, we identified obacunone as a candidate compound for ADPKD drug discovery from a natural antioxidant compound library. In vitro experiments showed that obacunone significantly inhibited cyst formation and expansion of MDCK cysts and embryonic kidney cysts in a dose-dependent manner. In vivo, obacunone treatment significantly reduced the renal cyst development in PKD mice. Western blot and morphological analysis revealed that obacunone served as a NRF2 activator in ADPKD, which suppressed lipid peroxidation by up-regulating GPX4 and finally restrained excessive cell proliferation by down-regulating mTOR and MAPK signaling pathways. Experimental data demonstrated obacunone as an effective renal cyst inhibitor for ADPKD, indicating that obacunone might be developed into a therapeutic drug for ADPKD treatment.
Collapse
|
8
|
Lu Y, Liu J, Tong A, Lu Y, Lv L. Interconversion and Acrolein-Trapping Capacity of Cardamonin/Alpinetin and Their Metabolites In Vitro and In Vivo. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:11926-11936. [PMID: 34587738 DOI: 10.1021/acs.jafc.1c04373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
People are at high risk of exposure to endogenous and exogenous acrolein (ACR). ACR can cause a multitude of illnesses, including cardiovascular disease, Alzheimer's disease, and diabetes. In this study, we investigated the reaction pathway of cardamonin (CAR) or alpinetin (ALP) with ACR and the interconversion of CAR and ALP in vitro at 37 °C in phosphate-buffered saline using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Subsequently, ACR adducts of CAR, ALP, and their metabolites, for example, CAR-ACR-1, ALP-ACR, mono-ACR-pinocembrin chalcone (PIN-ACR), and mono- and di-ACR-naringenin (NAR-ACR and NAR-2ACR), were detected in urine samples, but only CAR-ACR-1 and ALP-ACR were detected in fecal samples from the CAR- and ALP-treated mouse groups using ultraperformance liquid chromatography-MS/MS, respectively. Quantitative analyses showed that CAR, ALP, and their metabolites markedly scavenged ACR in a dose-dependent manner in vivo. Furthermore, we also found that the metabolites of CAR or ALP remained and promoted the ACR-trapping ability.
Collapse
Affiliation(s)
- Yang Lu
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, Jiangsu 210023, People's Republic of China
| | - Juan Liu
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, Jiangsu 210023, People's Republic of China
| | - Anqi Tong
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, Jiangsu 210023, People's Republic of China
| | - Yongling Lu
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, Jiangsu 210023, People's Republic of China
| | - Lishuang Lv
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Nanjing, Jiangsu 210023, People's Republic of China
| |
Collapse
|