1
|
Ge X, Liu T, Wang Y, Wen H, Huang Z, Chen L, Xu J, Zhou H, Wu Q, Zhao C, Shao R, Xu W. Porous starch microspheres loaded with luteolin exhibit hypoglycemic activities and alter gut microbial communities in type 2 diabetes mellitus mice. Food Funct 2025; 16:54-70. [PMID: 39377562 DOI: 10.1039/d4fo02907k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Luteolin (LUT), a natural flavonoid known for its hypoglycemic properties, is primarily sourced from vegetables such as celery and broccoli. However, its poor stability and low bioavailability in the upper digestive tract hinder its application in the functional food industry. To address these challenges, this study employed porous starch (PS) as a carrier to develop PS microspheres loaded with luteolin (PSLUT), simulating its release in vitro. The research assessed the hypoglycemic effects of LUT in type 2 diabetes mellitus (T2DM) mice both before and after PS treatment. In vitro findings demonstrated that PS improved LUT's stability in simulated gastric fluids and enhanced its in vivo bioavailability, aligning with experimental outcomes. PSLUT administration significantly improved body weight, fasting blood glucose (FBG), oral glucose tolerance test (OGTT), pancreatic islet function, and other relevant indicators in T2DM mice. Moreover, PSLUT alleviated abnormal liver biochemical indicators and liver tissue injury caused by T2DM. The underlying hypoglycemic mechanism of PSLUT is thought to involve the regulation of protein kinase B (AKT-1) and glucose transporter 2 (GLUT-2). After four weeks of intervention, various PSLUT doses significantly reduced the Firmicutes to Bacteroidetes ratio at the phylum level and decreased the relative abundance of harmful bacteria at the genus level, including Acetatifactor, Candidatus-Arthromitus, and Turicibacter. This microbial shift was associated with improvements in hyperglycemia-related indicators such as FBG, the area under the curve (AUC) of OGTT, and homeostasis model assessment of insulin resistance (HOMA-IR), which are closely linked to these bacterial genera. Additionally, Lachnoclostridium, Parasutterella, Turicibacter, and Papillibacter were identified as key intestinal marker genera involved in T2DM progression through Spearman correlation analysis. In conclusion, PS enhanced LUT's hypoglycemic efficacy by modulating the transcription and protein expression levels of AKT-1 and GLUT-2, as well as the relative abundance of potential gut pathogens in T2DM mice. These results provide a theoretical foundation for advancing luteolin's application in the functional food industry and further investigating its hypoglycemic potential.
Collapse
Affiliation(s)
- Xiaodong Ge
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, 224051, China.
| | - Tingting Liu
- Clinical Pharmacy Department, Yancheng Second People's Hospital, Yancheng, 224051, China
| | - Yaolin Wang
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, 224051, China.
| | - Huanhuan Wen
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, 224051, China.
| | - Zirui Huang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ligen Chen
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, 224051, China.
| | - Jianda Xu
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, Changzhou, 213003, China
| | - Hongcheng Zhou
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng, 224051, China
| | - Qin Wu
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng, 224051, China
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Rong Shao
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, 224051, China.
| | - Wei Xu
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, 224051, China.
| |
Collapse
|
2
|
Ambele MA, Maebele LT, Mulaudzi TV, Kungoane T, Damane BP. Advances in nano-delivery of phytochemicals for glioblastoma treatment. DISCOVER NANO 2024; 19:216. [PMID: 39718730 DOI: 10.1186/s11671-024-04172-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Glioblastoma (GBM) is an aggressive brain tumor characterized by cellular and molecular diversity. This diversity presents significant challenges for treatment and leads to poor prognosis. Surgery remains the primary treatment of choice for GBMs, but it often results in tumor recurrence due to complex interactions between GBM cells and the peritumoral brain zone. Phytochemicals have shown promising anticancer activity in in-vitro studies and are being investigated as potential treatments for various cancers, including GBM. However, some phytochemicals have failed to translate their efficacy to pre-clinical studies due to limited penetration into the tumor microenvironment, leading to high toxicity. Thus, combining phytochemicals with nanotechnology has emerged as a promising alternative for treating GBM. This review explores the potential of utilizing specific nanoparticles to deliver known anticancer phytochemicals directly to tumor cells. This method has demonstrated potential in overcoming the challenges of the complex GBM microenvironment, including the tight blood-brain barrier while minimizing damage to healthy brain tissue. Therefore, employing this interdisciplinary approach holds significant promise for developing effective phyto-nanomedicines for GBM and improving patient outcomes.
Collapse
Affiliation(s)
- Melvin Anyasi Ambele
- Department of Oral and Maxillofacial Pathology, Faculty of Health Sciences, School of Dentistry, University of Pretoria, P.O. Box 1266, Pretoria, 0001, South Africa.
- Department of Immunology, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, University of Pretoria, P.O. Box 0084, Gezina, South Africa.
| | - Lorraine Tshegofatso Maebele
- Department of Surgery, Level 7, Bridge E, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa
| | - Thanyani Victor Mulaudzi
- Department of Surgery, Level 7, Bridge E, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa
| | - Tsholofelo Kungoane
- Department of Oral and Maxillofacial Pathology, Faculty of Health Sciences, School of Dentistry, University of Pretoria, P.O. Box 1266, Pretoria, 0001, South Africa
| | - Botle Precious Damane
- Department of Surgery, Level 7, Bridge E, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa.
| |
Collapse
|
3
|
Eryilmaz IE, Colakoglu Bergel C, Arioz B, Huriyet N, Cecener G, Egeli U. Luteolin induces oxidative stress and apoptosis via dysregulating the cytoprotective Nrf2-Keap1-Cul3 redox signaling in metastatic castration-resistant prostate cancer cells. Mol Biol Rep 2024; 52:65. [PMID: 39699825 DOI: 10.1007/s11033-024-10178-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND The treatment of metastatic castration-resistant prostate cancer (mCRPC) is still challenging clinically. Due to the refractor and highly metastatic phenotype of mCRPC, novel therapy strategies need to be investigated. Luteolin, a promising anticancer agent with various biological targets in many cancer types, also has a pro-oxidant effect that selectively triggers ROS and apoptosis. In recent years, among its ROS-mediated mechanisms, the inhibitory effect of luteolin on the nuclear factor-E2-related factor 2 (Nrf2), the main ROS scavenger protein in cancer cells, has been reported. However, no evidence exists that luteolin potentially regulates the Nrf2 or its regulator signaling pathway, Nrf2-Keap1-Cul3 axis, concerning its pro-oxidant effects associated with ROS-triggered apoptosis in any PCa cells or tumor model. METHODS AND RESULTS In the present study, we investigated for the first time whether the anticancer effect of luteolin is associated with pro-oxidant activity via the regulation of the Nrf2-Keap1-Cul3 redox signaling in PC3 and DU145 mCRPC cells. The results showed that luteolin significantly caused more cytotoxic, apoptotic, and pro-oxidant effects in a dose-dependent manner in mCRPC cells than in WPMY-1 normal prostate fibroblast cells for 72 h. Moreover, significant inhibition of Nrf2-Keap1-Cul3 redox signaling has occurred in response to increasing doses of luteolin in mCRPC cells. CONCLUSIONS The current study put forth the potential pro-oxidant inhibitory effect of luteolin on the Nrf2-Keap1-Cul3 axis in mCRPC cells for the first time. Thus, luteolin might be an attractive therapy strategy with an inhibitory effect on the cytoprotective Nrf2-Keap1-Cul3 redox signaling for treating mCRPC.
Collapse
Affiliation(s)
- Isil Ezgi Eryilmaz
- Medical Biology Department, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.
| | | | - Bilge Arioz
- Medical Biology Department, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Nuseybe Huriyet
- Medical Biology Department, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Gulsah Cecener
- Medical Biology Department, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Unal Egeli
- Medical Biology Department, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
4
|
Godiyal Y, Maheshwari D, Taniguchi H, Zinzuwadia SS, Morera-Díaz Y, Tewari D, Bishayee A. Role of PD-1/PD-L1 signaling axis in oncogenesis and its targeting by bioactive natural compounds for cancer immunotherapy. Mil Med Res 2024; 11:82. [PMID: 39690423 DOI: 10.1186/s40779-024-00586-9] [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: 07/08/2024] [Accepted: 11/29/2024] [Indexed: 12/19/2024] Open
Abstract
Cancer is a global health problem and one of the leading causes of mortality. Immune checkpoint inhibitors have revolutionized the field of oncology, emerging as a powerful treatment strategy. A key pathway that has garnered considerable attention is programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1). The interaction between PD-L1 expressed on tumor cells and PD-1 reduces the innate immune response and thus compromises the capability of the body's immune system. Furthermore, it controls the phenotype and functionality of innate and adaptive immune components. A range of monoclonal antibodies, including avelumab, atezolizumab, camrelizumab, dostarlimab, durvalumab, sinitilimab, toripalimab, and zimberelimab, have been developed for targeting the interaction between PD-1 and PD-L1. These agents can induce a broad spectrum of autoimmune-like complications that may affect any organ system. Recent studies have focused on the effect of various natural compounds that inhibit immune checkpoints. This could contribute to the existing arsenal of anticancer drugs. Several bioactive natural agents have been shown to affect the PD-1/PD-L1 signaling axis, promoting tumor cell apoptosis, influencing cell proliferation, and eventually leading to tumor cell death and inhibiting cancer progression. However, there is a substantial knowledge gap regarding the role of different natural compounds targeting PD-1 in the context of cancer. Hence, this review aims to provide a common connection between PD-1/PD-L1 blockade and the anticancer effects of distinct natural molecules. Moreover, the primary focus will be on the underlying mechanism of action as well as the clinical efficacy of bioactive molecules. Current challenges along with the scope of future research directions targeting PD-1/PD-L1 interactions through natural substances are also discussed.
Collapse
Affiliation(s)
- Yogesh Godiyal
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Drishti Maheshwari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Hiroaki Taniguchi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552, Magdalenka, Poland
- African Genome Center, Mohammed VI Polytechnic University, Hay Moulay Rachid, 43150, Ben Guerir, Morocco
| | - Shweta S Zinzuwadia
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Yanelys Morera-Díaz
- Clinical Investigation and Biomedical Research Directions, Center for Genetic Engineering and Biotechnology, 11600, Havana, Cuba
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
5
|
Wang J, Li H, Wang Z, Ruan S. Luteolin: A Comprehensive and Visualized Analysis of Research Hotspots and Its Antitumor Mechanisms. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2377-2401. [PMID: 39686791 DOI: 10.1142/s0192415x24500903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The aim of this study was to analyze the research hotspots and mechanisms of luteolin in tumor-related fields using bibliometric and bioinformatic approaches to guide future research. We conducted a comprehensive screening of all articles on luteolin and tumors in Web of Science from 2008 to 2023. The extracted words from these publications were visualized using VOSviewer, Scimago Graphica, and CiteSpace. Public databases were used to collect luteolin and tumor-related targets. GO and KEGG analyses of luteolin antitumor-related genes were performed using Metascape. Protein interaction networks were built with Cytoscape and STRING, identifying hub targets of luteolin in antitumor activity. Subsequently, the binding capacity of luteolin to these hub targets was assessed using molecular docking technology. We found that China dominated this field, the Egyptian Knowledge Bank published the most articles, and Molecules had the highest number of related publications. Recently, network pharmacology, target, traditional Chinese medicine, and nanoparticles have become research hotspots in luteolin's antitumor research. A total of 483 overlapping genes between luteolin and tumors were identified, and they were closely associated with the cancer-associated pathways, PI3K-Akt, and MAPK signaling pathways. Luteolin forms a complex network of antitumor-related genes, with TP53, TNF, STAT3, AKT1, JUN, IL6, and SRC playing key roles and showing strong binding affinity to luteolin. Computer technology is becoming increasingly integral to the discipline, and future research will focus on more precise antitumor mechanisms and effective clinical applications.
Collapse
Affiliation(s)
- Jiaxuan Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| | - Hao Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| | - Zhenru Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| | - Shanming Ruan
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| |
Collapse
|
6
|
Yang Y, Chen Y, Jia X, Huang X. Association of dietary flavonoid intake with the prevalence and all-cause mortality of depressive symptoms: Findings from analysis of NHANES. J Affect Disord 2024; 366:44-58. [PMID: 39187180 DOI: 10.1016/j.jad.2024.08.150] [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: 06/21/2023] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND This study aimed to explore the relationship between flavonoids intake and the prevalence and all-cause mortality of depressive symptoms in American adults. METHODS Analyzing 2007-2008, 2009-2010, and 2017-2018 NHANES data, we examined the association between dietary flavonoid and depressive symptoms, including specific subclasses assessment and mortality outcomes tracking until December 31, 2019. Our methodology included weighted multivariate logistic regression, weighted cox proportional hazards regression and restricted cubic spline (RCS) models, supported by stratified and sensitivity analyses. RESULTS Among the 12,340 participants in total, 1129 exhibited depressive symptoms. The multiple logistic regression analysis showed a significant reduction in total flavonoid and subclass intake in individuals with current depressive symptoms. Adjusted odds ratios (ORs) for the highest quartiles were 0.69 for anthocyanidins and 0.63 for flavones. Interaction effects emerged in non-hypertensive, higher-income, and normal-weight groups for flavones intake. The RCS model indicated an L-shaped association between depressive symptoms and total flavonoid intake, with inflection points at 346 mg/day. During a median follow-up of 119 months, 148 deaths occurred among patients with depressive symptoms. Hazard ratios (HRs) for all-cause mortality showed a significant positive correlation between total flavonoid intake and survival in model 1 (HR = 0.56), with an optimal intake range of 45.2-948.3 mg/day according to the RCS model. LIMITATIONS The study relied on U.S. population survey data, potentially limiting generalizability. Unmeasured confounding factors may exist, and genetic factors were not considered. CONCLUSIONS Adequate intake of flavonoids, especially anthocyanidins and flavones, is associated with reduced odds of depressive symptoms. Additionally, optimal intake ranges of flavonoid intake for mental health benefits were observed for all-cause mortality in population with depressive symptoms.
Collapse
Affiliation(s)
- Yaqin Yang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yuemei Chen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaotong Jia
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyan Huang
- Department of Nephrology, Hunan Provincial Hospital of Chinese Medicine, Hengyang, China.
| |
Collapse
|
7
|
Demir K, Turgut R, Şentürk S, Işıklar H, Günalan E. The Therapeutic Effects of Bioactive Compounds on Colorectal Cancer via PI3K/Akt/mTOR Signaling Pathway: A Critical Review. Food Sci Nutr 2024; 12:9951-9973. [PMID: 39723045 PMCID: PMC11666977 DOI: 10.1002/fsn3.4534] [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: 07/04/2024] [Revised: 09/25/2024] [Accepted: 09/28/2024] [Indexed: 12/28/2024] Open
Abstract
Understanding the molecular signaling pathways of colorectal cancer (CRC) can be accepted as the first step in treatment strategy. Permanent mTOR signaling activation stimulates the CRC process via various biological processes. It supplies the survival of CRC stem cells, tumorigenesis, morbidity, and decreased response to drugs in CRC pathogenesis. Therefore, inhibition of the mTOR signaling by numerous bioactive components may be effective against CRC. The study aims to discuss the therapeutic capacity of various polyphenols, terpenoids, and alkaloids on CRC via the PI3K/Akt/mTOR pathway. The potential molecular effects of bioactive compounds on the mTOR pathway's upstream and downstream targets are examined. Each bioactive component causes various physiological processes, such as triggering free radical production, disruption of mitochondrial membrane potential, cell cycle arrest, inhibition of CRC stem cell migration, and suppression of glycolysis through mTOR signaling inhibition. As a result, carcinogenesis is inhibited by inducing apoptosis and autophagy. However, it should be noted that studies are primarily in vitro dose-dependent treatment researchers. This study raises awareness about the role of phenolic compounds in treating CRC, contributing to their future use as anticancer agents. These bioactive compounds have the potential to be developed into food supplementation to prevent and treat various cancer types including CRC. This review has the potential to lead to further development of clinical studies. In the future, mTOR inhibition by applying several bioactive agents using advanced drug delivery systems may contribute to CRC treatment with 3D cell culture and in vivo clinical studies.
Collapse
Affiliation(s)
- Kübra Demir
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
- Faculty of Health Science, Department of Nutrition and DieteticsSabahattin Zaim UniversityIstanbulTürkiye
| | - Rana Turgut
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
| | - Selcen Şentürk
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
| | - Handan Işıklar
- Faculty of Medicine, Department of Internal MedicineYalova UniversityYalovaTürkiye
| | - Elif Günalan
- Faculty of Health Science, Department of Nutrition and DieteticsIstanbul Health and Technology UniversityIstanbulTürkiye
| |
Collapse
|
8
|
Chen J, Ye W. Molecular mechanisms underlying Tao-Hong-Si-Wu decoction treating hyperpigmentation based on network pharmacology, Mendelian randomization analysis, and experimental verification. PHARMACEUTICAL BIOLOGY 2024; 62:296-313. [PMID: 38555860 PMCID: PMC11632782 DOI: 10.1080/13880209.2024.2330609] [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: 10/23/2023] [Revised: 02/26/2024] [Accepted: 03/02/2024] [Indexed: 04/02/2024]
Abstract
CONTEXT Hyperpigmentation, a common skin condition marked by excessive melanin production, currently has limited effective treatment options. OBJECTIVE This study explores the effects of Tao-Hong-Si-Wu decoction (THSWD) on hyperpigmentation and to elucidate the underlying mechanisms. MATERIALS AND METHODS We employed network pharmacology, Mendelian randomization, and molecular docking to identify THSWD's hub targets and mechanisms against hyperpigmentation. The Cell Counting Kit-8 (CCK-8) assay determined suitable THSWD treatment concentrations for PIG1 cells. These cells were exposed to graded concentrations of THSWD-containing serum (2.5%, 5%, 10%, 15%, 20%, 30%, 40%, and 50%) and treated with α-MSH (100 nM) to induce an in vitro hyperpigmentation model. Assessments included melanin content, tyrosinase activity, and Western blotting. RESULTS ALB, IL6, and MAPK3 emerged as primary targets, while quercetin, apigenin, and luteolin were the core active ingredients. The CCK-8 assay indicated that concentrations between 2.5% and 20% were suitable for PIG1 cells, with a 50% cytotoxicity concentration (CC50) of 32.14%. THSWD treatment significantly reduced melanin content and tyrosinase activity in α-MSH-induced PIG1 cells, along with downregulating MC1R and MITF expression. THSWD increased ALB and p-MAPK3/MAPK3 levels and decreased IL6 expression in the model cells. DISCUSSION AND CONCLUSION THSWD mitigates hyperpigmentation by targeting ALB, IL6, and MAPK3. This study paves the way for clinical applications of THSWD as a novel treatment for hyperpigmentation and offers new targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jun Chen
- Department of Geriatrics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Wenyi Ye
- Department of Traditional Chinese Internal Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
9
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [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: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
10
|
Yang Y, He S, Wang W, Lu Y, Ren B, Dan C, Ji Y, Yu R, Ju X, Qiao X, Xiao Y, Cai J, Hong X. NIR-II Image-Guided Wound Healing in Hypoxic Diabetic Foot Ulcers: The Potential of Ergothioneine-Luteolin-Chitin Hydrogels. Macromol Rapid Commun 2024; 45:e2400528. [PMID: 39422630 DOI: 10.1002/marc.202400528] [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: 06/29/2024] [Revised: 08/18/2024] [Indexed: 10/19/2024]
Abstract
Hypoxic diabetic foot ulcers (HDFUs) pose a challenging chronic condition characterized by oxidative stress damage, bacterial infection, and persistent inflammation. This study introduces a novel therapeutic approach combining ergothioneine (EGT), luteolin (LUT), and quaternized chitosan oxidized dextran (QCOD) to address these challenges and facilitate wound healing in hypoxic DFUs. In vitro, assessments have validated the biosafety, antioxidant, and antimicrobial properties of the ergothioneine-luteolin-chitin (QCOD@EGT-LUT) hydrogel. Furthermore, near-infrared II (NIR-II) fluorescence image-guided the application of QCOD@EGT-LUT hydrogel in simulated HDFUs. Mechanistically, QCOD@EGT-LUT hydrogel modulates the diabetic wound microenvironment by reducing reactive oxygen species (ROS). In vivo studies demonstrated increased expression of angiogenic factors mannose receptor (CD206) and latelet endothelial cell adhesion molecule-1 (PECAM-1/CD31), coupled with decreased inflammatory factors tumor necrosis factor-α (TNF-α) and Interleukin-6 (IL-6), thereby promoting diabetic wound healing through up-regulation of transforming growth factor β-1 (TGF-β1).
Collapse
Affiliation(s)
- Yao Yang
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Tibet University, Lhasa, 850000, China
| | - Shengnan He
- Key Laboratory of Virology and Biosafety (CAS), Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| | - Wumei Wang
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Tibet University, Lhasa, 850000, China
| | - Yiwen Lu
- Hubei Engineering Centre of Natural Polymers-Based Medical Materials, College of Chemistry & Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Bingtao Ren
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Tibet University, Lhasa, 850000, China
| | - Ci Dan
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Tibet University, Lhasa, 850000, China
| | - Yang Ji
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Tibet University, Lhasa, 850000, China
| | - Rui Yu
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Tibet University, Lhasa, 850000, China
| | - Xinpeng Ju
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Tibet University, Lhasa, 850000, China
| | - Xue Qiao
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Tibet University, Lhasa, 850000, China
- Key Laboratory of Virology and Biosafety (CAS), Shenzhen Institute of Wuhan University, Shenzhen, 518057, China
| | - Yuling Xiao
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Tibet University, Lhasa, 850000, China
| | - Jie Cai
- Hubei Engineering Centre of Natural Polymers-Based Medical Materials, College of Chemistry & Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Xuechuan Hong
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Tibet University, Lhasa, 850000, China
| |
Collapse
|
11
|
Nechchadi H, Nadir Y, Benhssaine K, Alem C, Sellam K, Boulbaroud S, Berrougui H, Ramchoun M. Hypolipidemic activity of phytochemical combinations: A mechanistic review of preclinical and clinical studies. Food Chem 2024; 459:140264. [PMID: 39068825 DOI: 10.1016/j.foodchem.2024.140264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024]
Abstract
Hyperlipidemia, a condition characterized by elevated levels of lipids in the blood, poses a significant risk factor for various health disorders, notably cardiovascular diseases. Phytochemical compounds are promising alternatives to the current lipid-lowering drugs, which cause many undesirable effects. Based on in vivo and clinical studies, combining phytochemicals with other phytochemicals, prebiotics, and probiotics and their encapsulation in nanoparticles is more safe and effective for managing hyperlipidemia than monotherapy. To this end, the results obtained and the mechanisms of action of these combinations were examined in detail in this review.
Collapse
Affiliation(s)
- Habiba Nechchadi
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, 23000 Beni Mellal, Morocco.
| | - Youssef Nadir
- Laboratory of Biological Engineering, Faculty of Sciences and Techniques, University Sultan Moulay Slimane, 23000 Beni Mellal, Morocco
| | - Khalid Benhssaine
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, 23000 Beni Mellal, Morocco
| | - Chakib Alem
- Biochemistry of Natural Products Team, Faculty of Sciences and Techniques, Moulay Ismail University, 52000 Errachidia, Morocco
| | - Khalid Sellam
- Biology, Environment and Health Team, Faculty of sciences and Techniques, Moulay Ismail University, 52000 Errachidia, Morocco
| | - Samira Boulbaroud
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, 23000 Beni Mellal, Morocco
| | - Hicham Berrougui
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, 23000 Beni Mellal, Morocco
| | - Mhamed Ramchoun
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, 23000 Beni Mellal, Morocco
| |
Collapse
|
12
|
Ye Z, Yang S, Chen L, Yu W, Xia Y, Li B, Zhou X, Cheng F. Luteolin alleviated calcium oxalate crystal induced kidney injury by inhibiting Nr4a1-mediated ferroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 136:156302. [PMID: 39662099 DOI: 10.1016/j.phymed.2024.156302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND The global incidence of calcium oxalate (CaOx) kidney stones is rising, and effective treatments remain limited. Luteolin (Lut), a naturally flavonoid present in several plants, is recognized for its anti-inflammatory, anti-injury, and neuroprotective effects. However, its effects on CaOx kidney stone formation and the associated kidney damage are still unknown. OBJECTIVE Our study seeks to explore the therapeutic impact of Lut on kidney injury and renal fibrosis caused by CaOx crystal and to elucidate the underlying mechanisms. METHODS CaOx stone models were established in mice via intraperitoneal injection of glyoxylate (Gly, 100 mg/kg) for 12 days. Lut (50 mg/kg or 100 mg/kg) was administered intraperitoneally 7 days before and with the period of Gly treatment. Kidney function and histopathology changes in renal tissues were assessed. RNA sequencing was used to explore potential mechanisms between the model and Lut treatment groups. Molecular docking simulations evaluated the interaction between Lut and the downstream target Nr4a1. Moreover, Nr4a1 knockout mice and knockdown plasmids were used to validate the mechanism of Lut in the treatment of CaOx crystal-induced kidney injury. RESULTS Lut significantly mitigated kidney injury and renal fibrosis induced by CaOx crystal, as evidenced by improved kidney function, histopathology staining and Western blot analysis. Lut treatment also significantly inhibited lipid peroxidation and mitochondrial injury. In vitro experiments further demonstrated that Lut treatment alleviated injury and fibrosis in HK-2 cells. Mechanistically, RNA sequencing and molecular docking simulations indicated that Lut binds to Nr4a1 to regulate ferroptosis, thereby alleviating kidney injury induced by CaOx crystal. Overexpression of Nr4a1 negated Lut's beneficial effects, whereas Nr4a1 knockout exhibited a protective effect against kidney injury. CONCLUSION Lut exerts its protective effects by inhibiting ferroptosis via targeting Nr4a1-Slc7a11-GPX4 pathway, alleviating kidney injury and renal fibrosis caused by CaOx crystal deposition.
Collapse
Affiliation(s)
- Zehua Ye
- Department of Urology, Renmin hospital of Wuhan university, Wuhan, 430060
| | - Songyuan Yang
- Department of Urology, Renmin hospital of Wuhan university, Wuhan, 430060
| | - Lijia Chen
- Department of Urology, Renmin hospital of Wuhan university, Wuhan, 430060
| | - Weimin Yu
- Department of Urology, Renmin hospital of Wuhan university, Wuhan, 430060
| | - Yuqi Xia
- Department of Urology, Renmin hospital of Wuhan university, Wuhan, 430060
| | - Bojun Li
- Department of Urology, Renmin hospital of Wuhan university, Wuhan, 430060
| | - Xiangjun Zhou
- Department of Urology, Renmin hospital of Wuhan university, Wuhan, 430060
| | - Fan Cheng
- Department of Urology, Renmin hospital of Wuhan university, Wuhan, 430060.
| |
Collapse
|
13
|
Vazhappilly CG, Alsawaf S, Mathew S, Nasar NA, Hussain MI, Cherkaoui NM, Ayyub M, Alsaid SY, Thomas JG, Cyril AC, Ramadan WS, Chelakkot AL. Pharmacodynamics and safety in relation to dose and response of plant flavonoids in treatment of cancers. Inflammopharmacology 2024:10.1007/s10787-024-01581-1. [PMID: 39580755 DOI: 10.1007/s10787-024-01581-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/02/2024] [Indexed: 11/26/2024]
Abstract
Despite the recent advancements in developing bioactive nutraceuticals as anticancer modalities, their pharmacodynamics, safety profiles, and tolerability remain elusive, limiting their success in clinical trials. The failure of anticancer drugs in clinical trials can be attributed to the changes in drug clearance, absorption, and cellular responses, which alter the dose-response efficacy, causing adverse health effects. Flavonoids demonstrate a biphasic dose-response phenomenon exerting a stimulatory or inhibitory effect and often follow a U-shaped curve in different preclinical cancer models. A double-edged sword, bioflavonoids' antioxidant or prooxidant properties contribute to their hormetic behavior and facilitate redox homeostasis by regulating the levels of reactive oxygen species (ROS) in cells. Emerging reports suggest a need to discuss the pharmacodynamic broad-spectrum of plant flavonoids to improve their therapeutic efficacy, primarily to determine the ideal dose for treating cancer. This review discusses the dose-response effects of a few common plant flavonoids against some types of cancers and assesses their safety and tolerability when administered to patients. Moreover, we have emphasized the role of dietary-rich plant flavonoids as nutraceuticals in cancer treatment and prevention.
Collapse
Affiliation(s)
- Cijo George Vazhappilly
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE.
| | - Seba Alsawaf
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Shimy Mathew
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, UAE
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah, UAE
| | - Noora Ali Nasar
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Maheen Imtiaz Hussain
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Noor Mustapha Cherkaoui
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Mohammed Ayyub
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Serin Yaser Alsaid
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Joshua George Thomas
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Asha Caroline Cyril
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Wafaa S Ramadan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, UAE
| | | |
Collapse
|
14
|
Zheng Y, Meng L, Qu L, Zhao C, Wang L, Ma J, Liu C, Shou C. Co-targeting TMEM16A with a novel monoclonal antibody and EGFR with Cetuximab inhibits the growth and metastasis of esophageal squamous cell carcinoma. J Transl Med 2024; 22:1046. [PMID: 39563381 DOI: 10.1186/s12967-024-05830-3] [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: 08/16/2024] [Accepted: 10/30/2024] [Indexed: 11/21/2024] Open
Abstract
The chloride channel transmembrane protein 16A (TMEM16A) possesses a calcium-activated property linked to tumor-promoting malignant phenotype and electrophysiological stability. Numerous studies have shown that TMEM16A exhibits aberrant amplification in various squamous cell carcinomas such as esophageal squamous cell carcinoma (ESCC) and is correlated with unfavorable outcomes of ESCC patients. Therefore, TMEM16A is considered as a promising therapeutic target for ESCC. Because of its intricate structure, the development of therapeutic antibodies directed against TMEM16A has not been documented. In this study, we produced a series of novel monoclonal antibodies targeting TMEM16A and identified mT16#5 as an antibody capable of inhibiting ESCC cells migration, invasion and TMEM16A ion channel activity. Additionally, based on the validation that TMEM16A was positively correlated with expression of EGFR and the interaction between them, the mT16#5 exhibited a synergistic inhibitory effect on ESCC metastasis and growth when administered in combination with Cetuximab in vivo. In terms of mechanism, we found that mT16A#5 inhibited the phosphorylation of PI3K, AKT and JNK. These results highlight the anti-growth and anti-metastasis capacity of the combination of mT16A#5 and Cetuximab in the treatment of ESCC by targeting TMEM16A and EGFR, and provide a reference for combinational antibody treatment in ESCC.
Collapse
Affiliation(s)
- Yutian Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Department of Pathology, National Center for Children's Health (NCCH), Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
| | - Lin Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Like Qu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chuanke Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lixin Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jiayi Ma
- Beijing National Day School, Beijing, 100039, China
| | - Caiyun Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Chengchao Shou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
15
|
Wang X, Wu L, Luo D, He L, Wang H, Peng B. Mechanism of action of Salvia miltiorrhiza on avascular necrosis of the femoral head determined by integrated network pharmacology and molecular dynamics simulation. Sci Rep 2024; 14:28479. [PMID: 39558045 PMCID: PMC11574184 DOI: 10.1038/s41598-024-79532-7] [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: 07/19/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024] Open
Abstract
Avascular necrosis of the femoral head (ANFH) is a progressive, multifactorial, and challenging clinical condition that often leads to hip dysfunction and deterioration. The pathogenesis of ANFH is complex, and there is no foolproof treatment strategy. Although some pharmacologic and surgical treatments have been shown to improve ANFH, the associated side effects and poor prognosis are of concern. Therefore, there is an urgent need to explore therapeutic interventions with superior efficacy and safety to improve the quality of life of patients with ANFH. Salvia miltiorrhiza (SM), a traditional Chinese medicine with a long history, is widely used for the treatment of cardiovascular and musculoskeletal diseases due to its multiple pharmacological activities. However, the molecular mechanism of SM for the treatment of ANFH is still unclear. Therefore, this study aimed to explore the potential targets and mechanisms of SM for the treatment of ANFH using network pharmacology and molecular modeling techniques. By searching multiple databases, we screened 52 compounds and 42 common targets involved in ANFH therapy and identified dan-shexinkum d, cryptotanshinone, tanshinone iia, and dihydrotanshinlactone as key compounds. Based on the protein-protein interaction (PPI) network, TP53, AKT1, EGFR, STAT3, BCL2, IL6, and TNF were identified as core targets. Subsequent enrichment analysis revealed that these targets were mainly enriched in the AGE-RAGE, IL-17, and TNF pathways, which were mainly associated with inflammatory responses, apoptosis, and oxidative stress. In addition, molecular docking and 100 nanoseconds molecular dynamics (MD) simulations showed that the bioactive compounds of SM had excellent affinity and binding strength to the core targets. Among them, dan-shexinkum d possessed the lowest binding free energy (-215.874 kcal/mol and - 140.277 kcal/mol, respectively) for AKT1 and EGFR. These results demonstrated the multi-component, multi-target, and multi-pathway intervention mechanism of SM in the treatment of ANFH, which provided theoretical basis and clues for further experimental validation and development of anti-ANFH drugs.
Collapse
Affiliation(s)
- Xiangjin Wang
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Lijiao Wu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Dan Luo
- Basic Medical College of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Langyu He
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Hao Wang
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Bo Peng
- Department of Respiratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China.
| |
Collapse
|
16
|
Philip M, Karakka Kal AK, Subhahar MB, Karatt TK, Graiban FM, Ajeebsanu MM, Joseph M, Jose SV. Investigation Into the Equine Metabolism of Phosphodiesterase-4 Inhibitor Roflumilast for Potential Doping Control. Drug Test Anal 2024. [PMID: 39551487 DOI: 10.1002/dta.3822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024]
Abstract
The phosphodiesterase 4 (PDE4) inhibitors constitute a relatively modern class of medications that are known for inducing bronchodilation and exhibiting anti-inflammatory properties within the body. Due to these properties, there is concern regarding their potential misuse as performance-enhancing substances in competitive sports. This study delves into the metabolic conversion of roflumilast in thoroughbred horses following oral administration and in vitro experimentation using equine liver microsomes and Cunninghamella elegans. High-performance liquid chromatography coupled with a Q Exactive Orbitrap mass spectrometer (HPLC-HRMS) was employed for analysis. The investigation identified 10 metabolites of roflumilast, including six phase I and four phase II metabolites from in vivo studies, and 11 metabolites from in vitro studies, consisting of eight phase I and three phase II metabolites. The identified biotransformation products encompassed processes such as hydroxylation, chlorine substitution, methylation, N-oxide formation, and even the dissociation of methylenecyclopropane and difluoromethane. Furthermore, the study identified three glucuronic acid and one sulfonic acid conjugated phase II metabolites of the investigated drug candidate. The aforementioned findings contribute to the detection and comprehension of the unauthorized utilization of roflumilast in equestrian sports.
Collapse
Affiliation(s)
- Moses Philip
- Equine Forensic Unit, Central Veterinary Research Laboratory, Dubai, UAE
| | | | | | - Tajudheen K Karatt
- Equine Forensic Unit, Central Veterinary Research Laboratory, Dubai, UAE
| | | | | | - Marina Joseph
- Department of Bacteriology, Diagnostic Section, Central Veterinary Research Laboratory, Dubai, UAE
| | - Shantymol V Jose
- Department of Bacteriology, Diagnostic Section, Central Veterinary Research Laboratory, Dubai, UAE
| |
Collapse
|
17
|
N'do JYP, Paré D, Bondé L, Hilou A. Comparative phytochemical profile and biological activity of three Terminalia species as alternative antimicrobial therapies. Heliyon 2024; 10:e40159. [PMID: 39583820 PMCID: PMC11584580 DOI: 10.1016/j.heliyon.2024.e40159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 11/26/2024] Open
Abstract
Ethnopharmacological relevance Medicinal plants can help combat antibiotic resistance by providing novel, active molecules. Three plant species of the Terminalia genus are widely used in traditional medicine in the Mouhoun region for the treatment of cutaneous and respiratory diseases. Therefore, it is important to determine the ethnopharmacological potential of bark extracts from the trunks of these three Terminalia species. Aim of the study This study compared the phytochemical and biological activities of extracts from three Terminalia species to determine their ethnopharmacology. Materials and methods The medicinal properties of the extracts were assessed based on their ability to inhibit the growth of the following microorganisms: Staphylococcus aureus, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, Candida albicans, Candida krusei, Candida glabrata, and Candida tropicalis. The significant interest in these medicinal plants among the local communities were elucidated by their antioxidant properties and phytochemical composition, along with the detection key bioactive compounds. Major phytochemical groups and phenolic compounds were determined using high-performance liquid chromatography with a diode array detector. These phytochemical findings were validated by evaluating the antioxidant capacity of the extracts using DPPH, FRAP, and ABTS assays. Results Hydroethanolic, ethanolic, and hexane extracts from the bark of three Terminalia species inhibited the growth of both bacteria and fungi, as evidenced by their minimum inhibitory concentrations (MICs).The findings showed that Terminalia species were most effective against various tested bacteria and fungi, with MICs ranging from 0.1 to 6.25 mg/mL. Terminalia avicennioides, Terminalia macroptera, and Terminalia laxiflora extracts demonstrated 50 % inhibition of DPPH at concentrations ranging from 0.04 to 0.6 mg/mL. Phytochemical analysis revealed the presence of several families of chemical compounds, such as total phenolics and flavonoids. Phenolic compounds identified by HPLC in ethanolic extracts of T. avicennioides, such as isorhamnetin, quercetin, and ferulic acid, are recognised for their antimicrobial and antioxidant properties. Conclusion These findings establish an ethnobotany for these three Terminalia species, with their chromatographic characteristics facilitating the identification of key molecules of interest. The ethanolic extract of T. avicennioides can be used in phytomedicinal formulations against bacterial (P. aeruginosa and S. aureus) and fungal (C. albicans and C. glabrata) infections, both of which are recurrently recorded in certain skin and respiratory tract diseases.
Collapse
Affiliation(s)
- Jotham Yhi-pênê N'do
- Laboratory of Applied Biochemistry and Chemistry, Joseph Ki-Zerbo University, 03 BP 7021, Ouagadougou, 03, Burkina Faso
| | - Dramane Paré
- Laboratory of Applied Biochemistry and Chemistry, Joseph Ki-Zerbo University, 03 BP 7021, Ouagadougou, 03, Burkina Faso
| | - Loyapin Bondé
- Laboratory of Plant Biology and Ecology, University Joseph Ki-Zerbo, 03 BP 7021, Ouagadougou, 03, Burkina Faso, Burkina Faso
| | - Adama Hilou
- Laboratory of Applied Biochemistry and Chemistry, Joseph Ki-Zerbo University, 03 BP 7021, Ouagadougou, 03, Burkina Faso
| |
Collapse
|
18
|
Singh D, Shukla G. The multifaceted anticancer potential of luteolin: involvement of NF-κB, AMPK/mTOR, PI3K/Akt, MAPK, and Wnt/β-catenin pathways. Inflammopharmacology 2024:10.1007/s10787-024-01596-8. [PMID: 39543054 DOI: 10.1007/s10787-024-01596-8] [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: 09/30/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
Cancer is the predominant and major cause of fatality worldwide, based on the different types of cancer. There is a limitation in the current treatment. So we need better therapeutic agents that counteract the progression and development of malignant tumours. Plant-derived products are closely related and useful for human health. Luteolin is a polyphenolic flavonoid bioactive molecule that is present in various herbs, vegetables, fruits, and flowers and exhibits chemoprotective and pharmacological activity against different malignancies. To offer innovative approaches for the management of various cancers, we present a comprehensive analysis of the latest discoveries on luteolin. The aim is to inspire novel concepts for the development of advanced pharmaceuticals targeting cancer and search specifically targeted reviews and research articles published from January 1999 to January 2024 that investigated the application of luteolin in various cancer management. A thorough literature search utilizing the keywords "luteolin" "Signalling Pathway" "cancer" and nanoparticles was performed in the databases of Google Scholar, Web of Science, SCOPUS, UGC care list and PubMed. Through the compilation of existing research, we have discovered that luteolin possesses several therapeutic actions against various cancer via a signaling pathway involving the of NF-κB regulation, AMPK/mTOR, toll-like receptor, Nrf-2, PI3K/Akt MAPK and Wnt/β-catenin and their underlying mechanism of action has been well understood. This review intended to completely integrate crucial information on natural sources, biosynthesis, pharmacokinetics, signaling pathways, chemoprotective and therapeutic properties against various cancers, and nanoformulation of luteolin.
Collapse
Affiliation(s)
- Deepika Singh
- Faculty of Health Sciences, Department of Pharmaceutical Sciences, Shalom Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India.
| | - Gaurav Shukla
- Faculty of Health Sciences, Department of Pharmaceutical Sciences, Shalom Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India
| |
Collapse
|
19
|
Saez Lancellotti TE, Avena MV, Funes AK, Bernal-López MR, Gómez-Huelgas R, Fornes MW. Exploring the impact of lipid stress on sperm cytoskeleton: insights and prospects. Nat Rev Urol 2024:10.1038/s41585-024-00952-1. [PMID: 39528754 DOI: 10.1038/s41585-024-00952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
The decline in male fertility correlates with the global rise in obesity and dyslipidaemia, representing significant public health challenges. High-fat diets induce metabolic alterations, including hypercholesterolaemia, hepatic steatosis and atherosclerosis, with detrimental effects on testicular function. Testicular tissue, critically dependent on lipids for steroidogenesis, is particularly vulnerable to these metabolic disruptions. Excessive lipid accumulation within the testes, including cholesterol, triglycerides and specific fatty acids, disrupts essential sperm production processes such as membrane formation, maturation, energy metabolism and cell signalling. This leads to apoptosis, impaired spermatogenesis, and abnormal sperm morphology and function, ultimately compromising male fertility. During spermiogenesis, round spermatids undergo extensive reorganization, including the formation of the acrosome, manchette and specialized filamentous structures, which are essential for defining the final sperm cell shape. In this Perspective, we examine the impact of high-fat diets on the cytoskeleton of spermatogenic cells and its consequences to identify the mechanisms underlying male infertility associated with dyslipidaemia. Understanding these processes may facilitate the development of therapeutic strategies, such as dietary interventions or natural product supplementation, that aim to address infertility in men with obesity and hypercholesterolaemia. The investigation of cytoskeleton response to lipid stress extends beyond male reproduction, offering insights with broader implications.
Collapse
Affiliation(s)
- Tania E Saez Lancellotti
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina.
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina.
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.
| | - María V Avena
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Abi K Funes
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María-Rosa Bernal-López
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Ricardo Gómez-Huelgas
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel W Fornes
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
20
|
Treder-Rochna N, Mańkowska A, Kujawa W, Harciarek M. The effectiveness of olfactory training for chronic olfactory disorder following COVID-19: a systematic review. Front Hum Neurosci 2024; 18:1457527. [PMID: 39588055 PMCID: PMC11586678 DOI: 10.3389/fnhum.2024.1457527] [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: 06/30/2024] [Accepted: 10/07/2024] [Indexed: 11/27/2024] Open
Abstract
Background Chronic olfactory disorders are some of the most frequent post-COVID-19 presentations. Olfactory training (OT) is currently the most popular method used for treating post-viral olfactory dysfunction (PVOD). We evaluated the effect of olfactory training on the chronic olfactory disorders of patients infected with COVID-19. Methodology A systematic literature search was performed per PRISMA guidelines in PubMed, Scopus, Web of Science, EBSCOhost, and the Cochrane Library. Only patients with chronic olfactory disorders of 30 days or more were included. The primary outcome was the olfactory score at the end of follow-up. In all studies, improvement was defined as a positive change over time in the results of objective psychophysical olfactory tests. The most commonly used test was the Sniffin' Sticks. Typically, outcome measures involved comparing the mean olfactory scores. In the Sniffin' Sticks test, an improvement was also indicated by a change of 5.5 points or more in the Threshold, Discrimination, and Identification scores. Results Fourteen studies (1.596 participants) were included in this review. Among the included studies, up to 10 were RCTs. Nine studies assessed the combined effects of adjuvant therapy and olfactory training, while five studies assessed only OT. Conclusions In our assessment, olfactory training alone produces significant improvements in chronic olfactory dysfunctions. However, a combined therapy approach is essential to achieve more effective outcomes. Integrating olfactory training with adjuvants like CoUltraPEALut, Cerebrolysin, and oral Vitamin A has demonstrated substantial benefits in enhancing post-COVID-19 olfactory function. Strict adherence to the OT protocol and extending the duration of OT to 3 months or more significantly enhance treatment outcomes.
Collapse
Affiliation(s)
- Natalia Treder-Rochna
- Faculty of Social Sciences, Institute of Psychology, University of Gdansk, Gdansk, Poland
| | | | | | - Michał Harciarek
- Faculty of Social Sciences, Institute of Psychology, University of Gdansk, Gdansk, Poland
| |
Collapse
|
21
|
Zhang J, Li C, Li W, Shi Z, Liu Z, Zhou J, Tang J, Ren Z, Qiao Y, Liu D. Mechanism of luteolin against non-small-cell lung cancer: a study based on network pharmacology, molecular docking, molecular dynamics simulation, and in vitro experiments. Front Oncol 2024; 14:1471109. [PMID: 39582546 PMCID: PMC11582065 DOI: 10.3389/fonc.2024.1471109] [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: 07/26/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024] Open
Abstract
Introduction Luteolin, a naturally occurring flavonoid compound, demonstrates promising anti-cancer properties. However, its mechanism against non-small-cell lung cancer (NSCLC) remains unknown. This study employed network pharmacology, molecular docking, molecular dynamics simulation (MDS), and in vitro experiments to investigate the potential mechanisms by which luteolin against NSCLC. Methods Initially, the potential targets of luteolin and NSCLC-related targets were identified from public databases such as TCMSP, GeneCards, OMIM, DrugBank, and TTD. Subsequently, the protein-protein interaction (PPI) network screening and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted. The binding affinity and stability of luteolin with the core targets were assessed using molecular docking and MDS. Finally, the results were validated by in vitro experiments. Results A total of 56 luteolin targets and 2145 NSCLC-related targets were identified. Six core targets, TP53, EGFR, AKT1, TNF, JUN, and CASP3, were screened via the PPI network. The GO and KEGG analyses indicated that luteolin's activity against NSCLC potentially involves PI3K-Akt, NF-kappa B, and other signaling pathways. Molecular docking revealed that luteolin had high binding affinity with the core targets. MDS confirmed the stable interaction between luteolin and key proteins TP53 and AKT1. in vitro, luteolin significantly inhibited the proliferation and migration of A549 cells, while also inducing apoptosis. In addition, luteolin downregulated the expression of p-Akt (Ser473), MDM2, and Bcl-2 but upregulated the expression of p53 and Bax, which was consistent with the effect of LY294002. Conclusion Luteolin had a good anti-NSCLC effect, and the apoptosis-inducing effect might be related to the Akt/MDM2/p53 signaling pathway.
Collapse
Affiliation(s)
- Jihang Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
- Research Center for Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Changling Li
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Wenyi Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenpeng Shi
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenguo Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Junyu Zhou
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Tang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zixuan Ren
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yun Qiao
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Deshan Liu
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
22
|
Yu F, Wang G, Chen X, Zhang Y, Yang C, Hu H, Wei L. Luteolin alleviates cerebral ischemia/reperfusion injury by regulating cell pyroptosis. Open Med (Wars) 2024; 19:20241063. [PMID: 39507105 PMCID: PMC11538924 DOI: 10.1515/med-2024-1063] [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: 05/09/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 11/08/2024] Open
Abstract
Objective This study aimed to clarify the roles and underlying mechanisms of luteolin in the progression of cerebral ischemia/reperfusion injury (CIRI). Methods A mouse model of CIRI was established using the middle cerebral artery occlusion (MCAO) method, after which luteolin was administered. Subsequently, neuronal apoptosis and pyroptosis were measured and the brain tissues of each group were subjected to RNA sequencing. Results Luteolin alleviated MCAO-induced brain infarction, apoptosis, and pyroptosis. RNA sequencing identified 3,379, 2,777, and 3,933 differentially expressed genes (DEGs) in the MCAO vs sham, MCAO vs MCAO + luteolin, and MCAO + luteolin vs sham groups, respectively. The identified DEGs showed enrichment in multiple processes, including pattern specification, forebrain development, anion transport, leukocyte migration, regulation of cell-cell adhesion, and positive regulation of the response to external stimuli, as well as the calcium, PI3K-AKT, JAK-STAT, NF-kappa B, IL-17, cAMP, cGMP-PKG, and Wnt signaling pathways. In addition, Ccl2 and Angpt2 interacted more with the other top 30 DEGs with high interaction weights. Finally, RT-qPCR results showed that MCAO induction significantly up-regulated the expression of Stoml3, Eomes, and Ms4a15 and down-regulated Nms, Ttr, and Avpr1a; however, luteolin could partially reverse the expression caused by MCAO. Conclusion Luteolin can alleviate brain infarction, apoptosis, and pyroptosis in CIRI, and may improve MCAO-induced CIRI by targeting the identified DEGs and their enriched pathways.
Collapse
Affiliation(s)
- Fei Yu
- Department of Neurology, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Guangxue Wang
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xingyi Chen
- Department of Medical Department, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yanfei Zhang
- Department of Neurosurgery, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Cheng Yang
- Department of Neurosurgery, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hui Hu
- Department of Neurology, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Liang Wei
- Department of Neurosurgery, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| |
Collapse
|
23
|
Du S, Wang Z, Zhu H, Tang Z, Li Q. Flavonoids attenuate inflammation of HGF and HBMSC while modulating the osteogenic differentiation based on microfluidic chip. J Transl Med 2024; 22:992. [PMID: 39488714 PMCID: PMC11531701 DOI: 10.1186/s12967-024-05808-1] [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: 05/29/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND When inflammation occurs in periodontal tissues, a dynamic cellular crosstalk interacts between gingival fibroblasts and bone marrow mesenchymal stem cells (BMSCs), which plays a crucial role in the biological behaviour and differentiation of the cells. Recently, flavonoids are increasingly recognized for their therapeutic potential in modulating inflammation and osteogenic differentiation. Owing to their varied molecular structures and mechanisms, there are more needs that flavonoid compounds should be identified by extensive screening. However, current drug research mostly relies on static, single-type cell cultures. In this study, an innovative bionic microfluidic chip system tailored for both soft and hard tissues was developed to screen for flavonoids suitable for treating periodontitis. METHODS This study developed a microfluidic system that bionically simulates the soft and hard structures of periodontal tissues. Live/dead staining, reactive oxygen species (ROS) staining, and RT-qPCR analysis were employed. These techniques evaluated the effects of flavonoid compounds on the levels of inflammatory factors and ROS contents in HGF and HBMSC under LPS stimulation. Additionally, the impact of these compounds on osteogenic induction in HBMSC and the exploration of the underlying mechanisms were assessed. RESULTS The microfluidic chip used in this study features dual chambers separated by a porous membrane, allowing cellular signal communication via bioactive factors secreted by cells in both layers under perfusion. The inflammatory response within the chip under LPS stimulation was lower compared to individual static cultures of HGF and HBMSC. The selected flavonoids-myricetin, catechin, and quercetin-significantly reduced cellular inflammation, decreased ROS levels, and enhanced osteogenic differentiation of BMSCs. Additionally, fisetin, silybin, and icariside II also demonstrated favorable outcomes in reducing inflammation, lowering ROS levels, and promoting osteogenic differentiation through the Wnt/β-catenin pathway. CONCLUSIONS The bionic microfluidic chip system provides enhanced capabilities for drug screening and evaluation, delivering a more precise assessment of drug efficacy and safety compared to traditional in vitro methods. This study demonstrates the efficacy of flavonoids in influencing osteogenic processes in BMSCs primarily through the Wnt/β-catenin pathway. These results uncover the potential of flavonoids as therapeutic medicine for treating periodontitis, meriting further research and development.
Collapse
Affiliation(s)
- Sa Du
- Second Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
- Center for Digital Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Zhongyu Wang
- Second Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Huilin Zhu
- Second Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Zhihui Tang
- Second Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China.
| | - Qing Li
- Center for Digital Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China.
| |
Collapse
|
24
|
Zhai P, Ouyang XH, Yang ML, Lin L, Li JY, Li YM, Cheng X, Zhu R, Hu DS. Luteolin protects against myocardial ischemia/reperfusion injury by reducing oxidative stress and apoptosis through the p53 pathway. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:652-664. [PMID: 39343710 DOI: 10.1016/j.joim.2024.09.001] [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: 09/01/2023] [Accepted: 08/13/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVE Myocardial ischemia/reperfusion injury (MIRI) is an obstacle to the success of cardiac reperfusion therapy. This study explores whether luteolin can mitigate MIRI by regulating the p53 signaling pathway. METHODS Model mice were subjected to a temporary surgical ligation of the left anterior descending coronary artery, and administered luteolin. The myocardial infarct size, myocardial enzyme levels, and cardiac function were measured. Latent targets and signaling pathways were screened using network pharmacology and molecular docking. Then, proteins related to the p53 signaling pathway, apoptosis and oxidative stress were measured. Hypoxia/reoxygenation (HR)-incubated HL1 cells were used to validate the effects of luteolin in vitro. In addition, a p53 agonist and an inhibitor were used to investigate the mechanism. RESULTS Luteolin reduced the myocardial infarcted size and myocardial enzymes, and restored cardiac function in MIRI mice. Network pharmacology identified p53 as a hub target. The bioinformatic analyses showed that luteolin had anti-apoptotic and anti-oxidative properties. Additionally, luteolin halted the activation of p53, and prevented both apoptosis and oxidative stress in myocardial tissue in vivo. Furthermore, luteolin inhibited cell apoptosis, JC-1 monomer formation, and reactive oxygen species elevation in HR-incubated HL1 cells in vitro. Finally, the p53 agonist NSC319726 downregulated the protective attributes of luteolin in the MIRI mouse model, and both luteolin and the p53 inhibitor pifithrin-α demonstrated a similar therapeutic effect in the MIRI mice. CONCLUSION Luteolin effectively treats MIRI and may ameliorate myocardial damage by regulating apoptosis and oxidative stress through its targeting of the p53 signaling pathway. Please cite this article as: Zhai P, Ouyang XH, Yang ML, Lin L, Li JY, Li YM, Cheng X, Zhu R, Hu DS. Luteolin protects against myocardial ischemia/reperfusion injury by reducing oxidative stress and apoptosis through the p53 pathway. J Integr Med. 2024; 22(6): 652-664.
Collapse
Affiliation(s)
- Pan Zhai
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Xiao-Hu Ouyang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Meng-Ling Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Lan Lin
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Jun-Yi Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Yi-Ming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Rui Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - De-Sheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China; China-Russia Medical Research Center for Stress Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| |
Collapse
|
25
|
Li Q, Tong Y, Chen J, Xie T. Targeting programmed cell death via active ingredients from natural plants: a promising approach to cancer therapy. Front Pharmacol 2024; 15:1491802. [PMID: 39584140 PMCID: PMC11582395 DOI: 10.3389/fphar.2024.1491802] [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: 09/05/2024] [Accepted: 10/09/2024] [Indexed: 11/26/2024] Open
Abstract
Cancer is a serious public health problem in humans, and prevention and control strategies are still necessary. Therefore, the development of new therapeutic drugs is urgently needed. Targeting programmed cell death, particularly via the induction of cancer cell apoptosis, is one of the cancer treatment approaches employed. Recently, an increasing number of studies have shown that compounds from natural plants can target programmed cell death and kill cancer cells, laying the groundwork for use in future anticancer treatments. In this review, we focus on the latest research progress on the role and mechanism of natural plant active ingredients in different forms of programmed cell death, such as apoptosis, autophagy, necroptosis, ferroptosis, and pyroptosis, to provide a strong theoretical basis for the clinical development of antitumor drugs.
Collapse
Affiliation(s)
- Qian Li
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yan Tong
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jianxiang Chen
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Tian Xie
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
26
|
Mod Razif MRF, Chan SY, Chew YL, Hassan M, Ahmad Hisham S, Abdul Rahman S, Mai CW, Teo MYM, Kee PE, Khoo KS, Lee SK, Liew KB. Recent Developments in Luteolin-Loaded Nanoformulations for Enhanced Anti-Carcinogenic Activities: Insights from In Vitro and In Vivo Studies. SCI 2024; 6:68. [DOI: 10.3390/sci6040068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2025] Open
Abstract
With approximately 18 million people affected by cancer in 2020 globally, scientists are exploring innovative approaches to develop effective treatments for various types of cancer. Traditional chemotherapy drugs, although effective against cancer cells, often lead to significant side effects on healthy tissues, such as hair loss, anemia, and nausea. To discover safer alternatives, researchers are investigating natural bioactive compounds found abundantly in plants. Luteolin, a flavonoid found in celery and artichokes, stands out due to its diverse anti-carcinogenic properties, including inhibiting proliferation, inducing apoptosis, activating autophagy, and inhibiting angiogenesis and metastasis. However, the therapeutic potential of luteolin is hindered by challenges related to its bioavailability and solubility. This critical review explores the specific anti-carcinogenic properties of luteolin while analyzing the impact of its limited bioavailability and solubility on effectiveness. Additionally, it investigates the outcomes of encapsulating luteolin in nanoformulations, providing insights into potential strategies for enhancing its anti-carcinogenic effects. Finally, the review compares the efficacy of luteolin with and without nanoformulations. This review provides valuable insights into the potential of utilizing luteolin-loaded nanoformulations as a safer and more effective method for treating cancer, contributing to the ongoing efforts in improving cancer care and outcomes worldwide.
Collapse
Affiliation(s)
| | - Siok Yee Chan
- School of Pharmaceutical Science, Universiti Sains Malaysia, Jalan Universiti, Gelugor 11700, PNG, Malaysia
| | - Yik-Ling Chew
- Faculty of Pharmaceutical Sciences, UCSI University, UCSI Heights, Jalan Puncak Menara Gading, Taman Connaught, Cheras 56000, KUL, Malaysia
| | - Masriana Hassan
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Jalan Universiti 1, Serdang 43400, SGR, Malaysia
| | - Shairyzah Ahmad Hisham
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya 63000, SGR, Malaysia
| | - Shamima Abdul Rahman
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya 63000, SGR, Malaysia
| | - Chun-Wai Mai
- Faculty of Pharmaceutical Sciences, UCSI University, UCSI Heights, Jalan Puncak Menara Gading, Taman Connaught, Cheras 56000, KUL, Malaysia
| | - Michelle Yee Mun Teo
- Faculty of Applied Sciences, UCSI University, UCSI Heights, Jalan Puncak Menara Gading, Taman Connaught, Cheras 56000, KUL, Malaysia
| | - Phei Er Kee
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chungli, Taoyuan 320, Taiwan
| | - Kuan Shiong Khoo
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chungli, Taoyuan 320, Taiwan
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, TN, India
| | - Siew-Keah Lee
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, Kajang 43000, SGR, Malaysia
| | - Kai Bin Liew
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya 63000, SGR, Malaysia
| |
Collapse
|
27
|
Choudhary MK, Pancholi B, Kumar M, Babu R, Garabadu D. A review on endoplasmic reticulum-dependent anti-breast cancer activity of herbal drugs: possible challenges and opportunities. J Drug Target 2024:1-26. [PMID: 39404107 DOI: 10.1080/1061186x.2024.2417189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Breast cancer (BC) is a major cause of cancer-related mortality across the globe and is especially highly prevalent in females. Based on the poor outcomes and several limitations of present management approaches in BC, there is an urgent need to focus and explore an alternate target and possible drug candidates against the target in the management of BC. The accumulation of misfolded proteins and subsequent activation of unfolded protein response (UPR) alters the homeostasis of endoplasmic reticulum (ER) lumen that ultimately causes oxidative stress in ER. The UPR activates stress-detecting proteins such as IRE1α, PERK, and ATF6, these proteins sometimes may lead to the activation of pro-apoptotic signaling pathways in cancerous cells. The ER stress-dependent antitumor activity could be achieved either through suppressing the adaptive UPR to make cells susceptible to ER stress or by causing chronic ER stress that may lead to triggering of pro-apoptotic signaling pathways. Several herbal drugs trigger ER-dependent apoptosis in BC cells. Therefore, this review discussed the role of fifty-two herbal drugs and their active constituents, focusing on disrupting the balance of the ER within cancer cells. Further, several challenges and opportunities have also been discussed in ER-dependent management in BC.Breast cancer (BC) is a major cause of cancer-related mortality across the globe and is especially highly prevalent in females. Based on the poor outcomes and several limitations of present management approaches in BC, there is an urgent need to focus and explore an alternate target and possible drug candidates against the target in the management of BC. The accumulation of misfolded proteins and subsequent activation of unfolded protein response (UPR) alters the homeostasis of endoplasmic reticulum (ER) lumen that ultimately causes oxidative stress in ER. The UPR activates stress-detecting proteins such as IRE1α, PERK, and ATF6, these proteins sometimes may lead to the activation of pro-apoptotic signaling pathways in cancerous cells. The ER stress-dependent antitumor activity could be achieved either through suppressing the adaptive UPR to make cells susceptible to ER stress or by causing chronic ER stress that may lead to triggering of pro-apoptotic signaling pathways. Several herbal drugs trigger ER-dependent apoptosis in BC cells. Therefore, this review discussed the role of fifty-two herbal drugs and their active constituents, focusing on disrupting the balance of the ER within cancer cells. Further, several challenges and opportunities have also been discussed in ER-dependent management in BC.
Collapse
Affiliation(s)
- Mayank Kumar Choudhary
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Bhaskaranand Pancholi
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Manoj Kumar
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Raja Babu
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Debapriya Garabadu
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
28
|
Wen L, Zhang W, Hu J, Chen T, Wang Y, Lv C, Li M, Wang L, Xiao F. Luteolin target HSPB1 regulates endothelial cell ferroptosis to protect against radiation vascular injury. PLoS One 2024; 19:e0311922. [PMID: 39392831 PMCID: PMC11469493 DOI: 10.1371/journal.pone.0311922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/26/2024] [Indexed: 10/13/2024] Open
Abstract
Vascular endothelial damage due to ionizing radiation is the main pathological process of radiation injury and the main cause of damage to various organs in nuclear accidents. Ferroptosis plays an important role in ionizing radiation-induced cell death. We have previously reported that luteolin is highly resistant to ferroptosis. In the present study, body weight, microvessel count, H&E, and Masson staining results showed that luteolin rescued radial vascular injury in vivo. Cell Counting Kit 8 (CCK8), Giemsa staining clarified the anti-ferroptosis ability of luteolin with low toxicity. Malondialdehyde (MDA), superoxide dismutase (SOD), NADP+/NADPH, Fe2+ staining, dihydroethidium (DHE) and MitoTracker assays for ferroptosis-related metrics, we found that luteolin enhances human umbilical vein endothelial cells (HUVECs) antioxidant damage capacity. Drug affinity responsive target stability (DARTS), surface plasmon resonance (SPR), computer simulated docking and western blot showed that heat shock protein beta-1 (HSPB1) is one of the targets of luteolin action. Luteolin inhibits ferroptosis by promoting the protein expression of HSPB1/solute carrier family 7 member 11 (SLC7A11)/ glutathione peroxidase 4 (GPX4). In conclusion, we have preliminarily elucidated the antioxidant damage ferroptosis ability and the target of action of luteolin to provide a theoretical basis for the application of luteolin in radiation injury diseases.
Collapse
Affiliation(s)
- Li Wen
- School of Nursing, Jilin University, Changchun, P. R. China
| | - Weiyuan Zhang
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, P. R. China
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, P. R. China
| | - Jia Hu
- Department of Cardiovascular, The Sixth Medical Center of Chinese PLA General Hospital, Haidian District, Beijing, China
| | - Tao Chen
- Department of Cardiovascular, The Sixth Medical Center of Chinese PLA General Hospital, Haidian District, Beijing, China
| | - Yiming Wang
- School of Nursing, Jilin University, Changchun, P. R. China
| | - Changchang Lv
- School of Nursing, Jilin University, Changchun, P. R. China
| | - Min Li
- Beijing Institute of Radiation Medicine, Beijing, P. R. China
| | - Lisheng Wang
- School of Nursing, Jilin University, Changchun, P. R. China
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, P. R. China
| | - Fengjun Xiao
- Beijing Institute of Radiation Medicine, Beijing, P. R. China
| |
Collapse
|
29
|
Hou M, John Martin JJ, Song Y, Wang Q, Cao H, Li W, Sun C. Dynamics of flavonoid metabolites in coconut water based on metabolomics perspective. FRONTIERS IN PLANT SCIENCE 2024; 15:1468858. [PMID: 39435019 PMCID: PMC11491327 DOI: 10.3389/fpls.2024.1468858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/10/2024] [Indexed: 10/23/2024]
Abstract
Coconut meat and coconut water have garnered significant attention for their richness in healthful flavonoids. However, the dynamics of flavonoid metabolites in coconut water during different developmental stages remain poorly understood. This study employed the metabolomics approach using liquid chromatography-tandem mass spectrometry (LC-MS/MS) to investigate the changes in flavonoid metabolite profiles in coconut water from two varieties, 'Wenye No.5'(W5) and Hainan local coconut (CK), across six developmental stages. The results showed that a total of 123 flavonoid metabolites including chalcones, dihydroflavonoids, dihydroflavonols, flavonoids, flavonols, flavonoid carboglycosides, and flavanols were identified in the coconut water as compared to the control. The total flavonoid content in both types of coconut water exhibited a decreasing trend with developmental progression, but the total flavonoid content in CK was significantly higher than that in W5. The number of flavonoid metabolites that differed significantly between the W5 and CK groups at different developmental stages were 74, 74, 60, 92, 40 and 54, respectively. KEGG pathway analysis revealed 38 differential metabolites involved in key pathways for flavonoid biosynthesis and secondary metabolite biosynthesis. This study provides new insights into the dynamics of flavonoid metabolites in coconut water and highlights the potential for selecting and breeding high-quality coconuts with enhanced flavonoid content. The findings have implications for the development of coconut-based products with improved nutritional and functional properties.
Collapse
Affiliation(s)
- Mingming Hou
- School of Life Sciences, Henan University, Kaifeng, Henan, China
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang, China
| | - Jerome Jeyakumar John Martin
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang, China
- National Key Laboratory for Tropical Crop Breeding, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Yuqiao Song
- School of Life Sciences, Henan University, Kaifeng, Henan, China
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang, China
- National Key Laboratory for Tropical Crop Breeding, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Qi Wang
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang, China
- College of Wine and Horticulture, Ningxia University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Hongxing Cao
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang, China
- National Key Laboratory for Tropical Crop Breeding, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Wenrao Li
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Chengxu Sun
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang, China
| |
Collapse
|
30
|
Bona NP, Pedra NS, Spohr L, da Silva Dos Santos F, Saraiva JT, Carvalho FB, da Cruz Fernandes M, Fernandes AS, Saraiva N, Martins MF, Tavares RG, Spanevello RM, Aguiar MSSD, Stefanello FM. Antitumoral Activity of Cecropia Pachystachya Leaves Extract in Vitro and in Vivo Model of Rat Glioma: Brain and Blood Effects. Mol Neurobiol 2024; 61:8234-8252. [PMID: 38483655 DOI: 10.1007/s12035-024-04086-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 03/02/2024] [Indexed: 09/21/2024]
Abstract
The aim of this study was to investigate the antiglioma effect of Cecropia pachystachya Trécul (CEC) leaves extract against C6 and U87 glioblastoma (GB) cells and in a rat preclinical GB model. The CEC extract reduced in vitro cell viability and biomass. In vivo, the extract decreased the tumor volume approximately 62%, without inducing systemic toxicity. The deficit in locomotion and memory and an anxiolytic-like behaviors induced in the GB model were minimized by CEC. The extract decreased the levels of reactive oxygen species, nitrites and thiobarbituric acid reactive substances and increased the activity of antioxidant enzymes in platelets, sera and brains of GB animals. The activity of NTPDases, 5'-nucleotidase and adenosine deaminase (ADA) was evaluated in lymphocytes, platelets and serum. In platelets, ATP and AMP hydrolysis was reduced and hydrolysis of ADP and the activity of ADA were increased in the control, while in CEC-treated animals no alteration in the hydrolysis of ADP was detected. In serum, the reduction in ATP hydrolysis was reversed by CEC. In lymphocytes, the increase in the hydrolysis of ATP, ADP and in the activity of ADA observed in GB model was altered by CEC administration. The observed increase in IL-6 and decrease in IL-10 levels in the serum of GB animals was reversed by CEC. These results demonstrate that CEC extract is a potential complementary treatment to GB, decreasing the tumor size, while modulating aspects of redox and purinergic systems.
Collapse
Affiliation(s)
- Natália Pontes Bona
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, 96010-900, Brazil
| | - Nathalia Stark Pedra
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Luiza Spohr
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Francieli da Silva Dos Santos
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, 96010-900, Brazil
| | - Juliane Torchelsen Saraiva
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, 96010-900, Brazil
| | - Fabiano Barbosa Carvalho
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Marilda da Cruz Fernandes
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Ana Sofia Fernandes
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa, 1749-024, Portugal
| | - Nuno Saraiva
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa, 1749-024, Portugal
| | - Marta Filipa Martins
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa, 1749-024, Portugal
- Department of Biomedical Sciences, University of Alcalá, Ctra, Madrid-Barcelona Km. 33.600, Alcalá de Henares, Madrid, 28871, Spain
| | - Rejane Giacomelli Tavares
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa, 1749-024, Portugal
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Roselia Maria Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Mayara Sandrielly Soares de Aguiar
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Francieli Moro Stefanello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, 96010-900, Brazil.
| |
Collapse
|
31
|
Lei W, Li X, Li S, Zhou F, Guo Y, Zhang M, Jin X, Zhang H. Targeting neutrophils extracellular traps, a promising anti-thrombotic therapy for natural products from traditional Chinese herbal medicine. Biomed Pharmacother 2024; 179:117310. [PMID: 39226727 DOI: 10.1016/j.biopha.2024.117310] [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: 05/13/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 09/05/2024] Open
Abstract
Thrombi are the main cause of vascular occlusion and contribute significantly to cardiovascular events and death. Neutrophils extracellular traps (NETs)-induced thrombosis plays a vital role in thrombotic complications and it takes the main responsibility for the resistance of fibrinolysis. However, the conventional anti-thrombotic therapies are inadequate to treat NETs-induced thrombotic complications but carry a high risk of bleeding. Consequently, increased attention has shifted towards exploring novel anti-thrombotic treatments targeting NETs. Interestingly, accumulating evidences prove that natural products from traditional Chinese herbal medicines have a great potential to mitigate thrombosis through inhibiting generous NETs formation and degrading excessive NETs. In this review, we elaborated the formation and degradation of NETs and highlighted its pivotal role in immunothrombosis through interactions with platelets and coagulation factors. Since available anti-thrombotic drugs targeting NETs are deficient, we further summarized the natural products and compounds from traditional Chinese herbal medicines which exert effective actions on regulating NETs formation and also have anti-thrombotic effects. Our findings underscore the diverse effects of natural products in targeting NETs, including relieving inflammation and oxidative stress of neutrophils, inhibiting neutrophils activation and DNA efflux, suppressing granule proteins release, reducing histones and promoting DNA degradation. This review aims to highlight the significance of natural medicines in anti-thrombotic therapies through targeting NETs and to lay a groundwork for developing novel anti-thrombotic agents from traditional Chinese herbal medicines.
Collapse
Affiliation(s)
- Wei Lei
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine,10 Poyanghu Road, Jinghai District, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiao Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine,10 Poyanghu Road, Jinghai District, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shanze Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine,10 Poyanghu Road, Jinghai District, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fengjie Zhou
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine,10 Poyanghu Road, Jinghai District, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yadi Guo
- School of Management, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Mingyan Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine,10 Poyanghu Road, Jinghai District, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xinyao Jin
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine,10 Poyanghu Road, Jinghai District, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine,10 Poyanghu Road, Jinghai District, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
32
|
Li YY, Chen YJ, Abdalbage Mohammed Abdalsadeg S, Xu KX, Ma LL, Moosavi-Movahedi AA, Hong J, Xiao BL. Biosensor Based on ZIF-67-HRP and MWCNTs Nanocomposite Modified Glass Carbon Electrode for the Detection of Luteolin in Vegetables. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:20495-20504. [PMID: 39287927 DOI: 10.1021/acs.langmuir.4c02037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Luteolin has various pharmacological properties, including anti-inflammatory, antioxidant, and antitumor characteristics. Due to its potential value in drugs and functional foods, it is important to develop an efficient method for detecting luteolin. In this work, the poor selectivity of existing luteolin nonenzymatic sensors was solved by translating the enzyme-catalyzed reaction from bulk solution to the surface of a horseradish peroxidase (HRP) modified electrode through an electrocatalytic oxidation process. Here, we modified the surface of a glassy carbon electrode (GCE) with metal-organic frameworks (MOFs; ZIF-67 here, abbreviated as ZIF), functional nanomaterials, and HRP and finally covered it with Nafion (NF). In this case, luteolin acts as a hydrogen donor, and the electrode acts as a hydrogen acceptor; the oxidation reaction occurs on the electrode surface. The use of ZIF-67 ensured the conformational stability of HRP to ensure the selectivity and anti-interference property, and SDS-dispersed multiwalled carbon nanotubes (MWCNTs) enhanced the electrode conductivity. The use of NF avoids shedding of the electrode material during the testing process. A UV-vis spectrophotometer was used to study the selectivity of luteolin by HRP and the compatibility between HRP and ZIF. The materials were characterized and analyzed by scanning electron microscopy and transmission electron microscopy. Due to the synergistic effect of these nanomaterials, the linear range of NF/ZIF-HRP/MWCNTs-SDS/GCE was 1.0 × 10-2 to 6.0 μM, with detection limits of 25.3 nM (S/N = 3). The biosensor showed long-term stability and reproducibility, with a relative standard deviation of 4.2% for the peak current (n = 5). Finally, the biosensor was successfully used to detect luteolin in carrots, celery, and cauliflower.
Collapse
Affiliation(s)
- Yu-Ying Li
- School of Life Sciences, Henan University, Kaifeng 475000, China
| | - Yu-Jie Chen
- School of Life Sciences, Henan University, Kaifeng 475000, China
| | | | - Ke-Xin Xu
- School of Life Sciences, Henan University, Kaifeng 475000, China
| | - Lin-Lin Ma
- School of Life Sciences, Henan University, Kaifeng 475000, China
| | | | - Jun Hong
- School of Life Sciences, Henan University, Kaifeng 475000, China
| | - Bao-Lin Xiao
- School of Life Sciences, Henan University, Kaifeng 475000, China
| |
Collapse
|
33
|
Meneses-Sagrero SE, Rascón-Valenzuela LA, Arellano-García ME, Toledano-Magaña Y, García-Ramos JC. Natural compounds combined with imatinib as promising antileukemic therapy: An updated review. Fitoterapia 2024; 178:106185. [PMID: 39142530 DOI: 10.1016/j.fitote.2024.106185] [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: 06/12/2024] [Revised: 08/02/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
Natural products (NP) have been an alternative therapy for several diseases for centuries, and they also serve as an essential source of bioactive molecules, enhancing our drug discovery capacity. Among these NP, some phytochemicals have shown multiple biological effects, including anticancer activity, with higher effectiveness and less toxicity than actual treatments, suggesting their possible use on resilient human malignancies such as leukemia. Imatinib mesylate (Im) is a selective tyrosine kinase inhibitor widely used as an anticancer drug, the gold standard to attend chronic myeloid leukemia (CML). Nevertheless, resistance to this drug in patients with CML renders it insufficient to eliminate cells with Philadelphia chromosome (BCR/ABL+). Moreover, recent studies show that imatinib can induce genotoxic and chromosomic damage in some in vitro and in vivo models. These facts urge finding new therapeutic alternatives to increase the effectiveness of antileukemic treatment. Recent research has shown that the combined effects of phytochemicals with imatinib can improve the cytotoxicity or resensitized the resistant cells to this drug in diverse leukemia cell lines. Independent mechanisms of action among phytochemicals and imatinib include BCR/ABL regulation, downregulation of transcription factors, inhibition of anti-apoptotic and activation of pro-apoptotic proteins, apoptosis induction dependent- and independent of ROS-overproduction, membrane functions disruption, induction of cell cycle arrest, and cell death. This review summarizes and discusses the synergic effect of some phytochemicals combined with imatinib on leukemia cells and the mechanism of action proposed for these combinations, looking to contribute to developing new effective alternatives for leukemia treatment.
Collapse
Affiliation(s)
| | - Luisa Alondra Rascón-Valenzuela
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora, Blvd. Luis Encinas y Rosales S/N, Col. Centro, Hermosillo, Sonora, Mexico
| | - María Evarista Arellano-García
- Facultad de Ciencias, Universidad Autónoma de Baja California, Ctra. Transpeninsular No. 3917, Col. Playitas, Ensenada, Baja California, Mexico
| | - Yanis Toledano-Magaña
- Instituto Tecnológico de Ensenada, Tecnológico Nacional de México, Blvd. Tecnológico #150, Ex Ejido Chapultepec, Ensenada, Baja California, Mexico; Centro de Bachillerato Tecnológico Industrial y de Servicios No. 41. Dirección General de Educación Tecnológica Industrial y de Servicios, Carr. Transpeninsular km 115, Ex-Ejido Chapultepec, Ensenada, Baja California, Mexico.
| | - Juan Carlos García-Ramos
- Instituto Tecnológico de Ensenada, Tecnológico Nacional de México, Blvd. Tecnológico #150, Ex Ejido Chapultepec, Ensenada, Baja California, Mexico; Centro de Bachillerato Tecnológico Industrial y de Servicios No. 41. Dirección General de Educación Tecnológica Industrial y de Servicios, Carr. Transpeninsular km 115, Ex-Ejido Chapultepec, Ensenada, Baja California, Mexico.
| |
Collapse
|
34
|
Ahn YR, Jang JY, Kang YJ, Oh HJ, Kang MK, Yoon D, Kim HS, Moon HR, Chung HY, Kim ND. MHY446 induces apoptosis via reactive oxygen species-mediated endoplasmic reticulum stress in HCT116 human colorectal cancer cells. J Chemother 2024; 36:483-500. [PMID: 38054850 DOI: 10.1080/1120009x.2023.2286757] [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: 09/07/2023] [Revised: 11/03/2023] [Accepted: 11/18/2023] [Indexed: 12/07/2023]
Abstract
This study investigated the potential of a newly synthesized histone deacetylase (HDAC) inhibitor, MHY446, in inducing cell death in HCT116 colorectal cancer cells and compared its activity with that of suberoylanilide hydroxamic acid (SAHA), a well-known HDAC inhibitor. The results showed that MHY446 increased the acetylation of histones H3 and H4 and decreased the expression and activity of HDAC proteins in HCT116 cells. Additionally, MHY446 was confirmed to bind more strongly to HDAC1 than HDAC2 and inhibit its activity. In vivo experiments using nude mice revealed that MHY446 was as effective as SAHA in inhibiting HCT116 cell-grafted tumor growth. This study also evaluated the biological effects of MHY446 on cell survival and death pathways. The reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC) confirmed that ROS play a role in MHY446-induced cell death by reducing poly(ADP-ribose) polymerase cleavage. MHY446 also induced cell death via endoplasmic reticulum (ER) stress by increasing the expression of ER stress-related proteins. NAC treatment decreased the expression of ER stress-related proteins, indicating that ROS mediate ER stress as an upstream signaling pathway and induce cell death. While MHY446 did not exhibit superior HDAC inhibition efficacy compared to SAHA, it is anticipated to provide innovative insights into the future development of therapeutic agents for human CRC by offering novel chemical structure-activity relationship-related information.
Collapse
Affiliation(s)
- Yu Ra Ahn
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Jung Yoon Jang
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Yong Jung Kang
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Hye Jin Oh
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Min Kyung Kang
- Department of Manufacturing Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Dahye Yoon
- Department of Manufacturing Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hyung Ryong Moon
- Department of Manufacturing Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Nam Deuk Kim
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
35
|
Yang L, Wu L, Li Y, Yang Y, Gu Y, Yang J, Zhang L, Meng F. Comprehensive Secondary Metabolite Profiling and Antioxidant Activity of Aqueous and Ethanol Extracts of Neolamarckia cadamba (Roxb.) Bosser Fruits. Metabolites 2024; 14:511. [PMID: 39330518 PMCID: PMC11434403 DOI: 10.3390/metabo14090511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Neolamarckia cadamba (Rubiaceae) is a well-recognized medicinal plant with recorded therapeutical attributes. However, a thorough assessment of active compounds in its fruits is lacking, limiting their use and valorization in pharmacological industries. METHODS Thus, this study investigated variations in the fruits' secondary metabolite (SM) profiles, as well as antioxidant activities in aqueous (WA) and ethanol (ET) extracts. RESULTS Liquid chromatography-electrospray ionization tandem mass spectrometry identified 541 SMs, of which 14 and 1 (di-O-glucosylquinic acid) were specifically detected in ET and WA, respectively. Phenolic acids (36.97%), flavonoids (28.10%), terpenoids (12.20%), and alkaloids (9.98%) were the dominant SMs. The SM profiles of the fruits in WA and ET were quite different. We revealed 198 differentially extracted (DE) metabolites between WA and ET, including 62 flavonoids, 57 phenolic acids, 45 terpenoids, 14 alkaloids, etc. Most DE flavones (36 out of 40), terpenoids (45 out of 45), and alkaloids (12 out of 14) had higher content in ET. Catechin and its derivatives, procyanidins, and tannins had higher content in WA. ABTS and DPPH assays showed that the antioxidant activity of ET was significantly higher than that of WA. CONCLUSIONS Our findings will facilitate the efficient extraction and evaluation of specific active compounds in N. cadamba.
Collapse
Affiliation(s)
- Lin Yang
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519040, China (L.W.)
| | - Liyan Wu
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519040, China (L.W.)
| | - Yongxin Li
- College of Food Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Yuhui Yang
- College of Food Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Yuting Gu
- College of Food Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Jialin Yang
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519040, China (L.W.)
- College of Life Science, Jilin University, Changchun 130000, China
| | - Luzy Zhang
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519040, China (L.W.)
| | - Fanxin Meng
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519040, China (L.W.)
| |
Collapse
|
36
|
Li R, Ke H, Liu P, Yang Q, Li Y, Ke L, Wang X, Wu C, Zhang Y. Mechanisms of Yiai Fuzheng formula in the treatment of triple-negative breast cancer based on UPLC-Q-Orbitrap-HRMS, network pharmacology, and experimental validation. Heliyon 2024; 10:e36579. [PMID: 39319146 PMCID: PMC11419912 DOI: 10.1016/j.heliyon.2024.e36579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/26/2024] Open
Abstract
Ethnopharmacological relevance Yiai Fuzheng formula (YAFZF), as a Traditional Chinese Medicine (TCM) prescription, has been used widely at Zhongnan Hospital of Wuhan University for its therapeutic effects and high safety on triple-negative breast cancer (TNBC). Objective In this study, we employed ultra-high-performance liquid chromatography-quadrupole/orbitrap high-resolution mass spectrometry (UPLC-Q-Orbitrap-HRMS), network pharmacology, and experimental validation to elucidate the underlying action mechanism of YAFZF in the treatment of TNBC. Methods The key active ingredients in YAFZF were analyzed using UPLC-Q-Orbitrap-HRMS, and then the potential components, target genes and signalling pathways of YAFZF were predicted using the network pharmacological method. We then used molecular docking to visualize the combination characteristics between major active components and macromolecules in the crucial pathway. In vitro experiments were conducted to investigate the inhibitory effects of YAFZF treatment on the cell viability, invasion, and migration of 4T1 and MDA-MB-231 cells. The xenograft TNBC models were constructed using female Balb/c mice, and their body weights, tumour volumes, and weights were monitored during YAFZF treatment. Quantitative real-time PCR (qRT-PCR), Hematoxylin-eosin (HE), immunohistochemistry (IHC) staining, Western blot (WB), and terminal deoxynucleotidyl transferase (TdT)-dUTP nick-end labeling (TUNEL) staining were used for further experimental validation. Results Based on UPLC-Q-Orbitrap-HRMS and network pharmacology analysis, 6 major bioactive components and 153 intersecting genes were obtained for YAFZF against TNBC. Functional enrichment analysis identified that the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt) signalling pathway might be the mechanism of action of YAFZF in the treatment of TNBC. Molecular docking results suggested that the main active compounds in YAFZF had strong binding energies with the proteins in the PI3K/Akt pathway. In vitro experiments showed that YAFZF inhibited the cell viability, invasion, and migration abilities of TNBC cells. Animal experiments confirmed that YAFZF treatment suppressed tumour cell proliferation and increased apoptotic cells. PCR, HE, WB, and IHC results indicated that YAFZF could suppress xenograft tumour metastases by inhibiting the PI3K/AKT/mTOR pathway regulating the epithelial-mesenchymal transition (EMT) process. Conclusion YAFZF therapy showed its potential for reducing proliferation, invasion, and migration abilities, increasing apoptosis of TNBC cells. Furthermore, YAFZF treated TNBC by inhibiting xenograft tumour distant metastases via the regulation of EMT by the PI3K/Akt/mTOR pathway, suggesting that it may be useful as an adjuvant treatment.
Collapse
Affiliation(s)
- Ruijie Li
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Haoliang Ke
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Pan Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Qian Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yuxin Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Longzhu Ke
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiuping Wang
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Chaoyan Wu
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yingwen Zhang
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| |
Collapse
|
37
|
Dwikarina A, Bayati M, Efrat N, Roy A, Lei Z, Ho KV, Sumner L, Greenlief M, Thomas AL, Applequist W, Townesmith A, Lin CH. Exploring American Elderberry Compounds for Antioxidant, Antiviral, and Antibacterial Properties Through High-Throughput Screening Assays Combined with Untargeted Metabolomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.611920. [PMID: 39314315 PMCID: PMC11419141 DOI: 10.1101/2024.09.13.611920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
American elderberry (Sambucus nigra subsp. canadensis) is a rapidly emerging new perennial crop for Missouri, recognized for its high level of bioactive compounds with significant health benefits, including antibacterial, antiviral, and antioxidant properties. A high-throughput screening assay combined with untargeted metabolomics analysis was utilized on American elderberry juice from 21 genotypes to explore and characterize these bioactive compounds. Our metabolomics study has identified 32 putative bioactive compounds in the American Elderberry juices. An array of high-throughput screening bioassays was conducted to evaluate 1) total antioxidant capacity, 2) activation of antioxidant response elements (ARE), 3) antiviral activity, and 4) antibacterial activity of the putatively identified compounds. Our results revealed that 14 of the 32 American elderberry compounds exhibited strong antioxidant activity. Four compounds (isorhamnetin 3-O-glucoside, kaempferol, quercetin, and naringenin) activated ARE activity and were found to be non-cytotoxic to cells. Notably, six of the 32 compounds demonstrated significant antiviral activity in an in vitro TZM-bl assay against two strains of HIV-1 virus, CXCR4-dependent NL4-3 virus and CCR5-dependent BaL virus. Luteolin showed the most potent anti-HIV activity against the NL4-3 virus (IC50 = 1.49 μM), followed by isorhamnetin (IC50 = 1.67 μM). The most potent anti-HIV compound against the BaL virus was myricetin (IC50 = 1.14 μM), followed by luteolin (IC50 = 4.38 μM). Additionally, six compounds were found to have antibacterial activity against gram-positive bacteria S. aureus, with cyanidin 3-O-rutinoside having the most potent antibacterial activity in vitro (IC50 = 2.9 μM), followed by cyanidin 3-O-glucoside (IC50 = 3.7 μM). These findings support and validate the potential health benefits of compounds found in American elderberry juices and highlight their potential for use in dietary supplements as well as innovative applications in health and medicine.
Collapse
Affiliation(s)
- Amanda Dwikarina
- Center for Agroforestry, University of Missouri, Columbia, MO, U.S.A
- School of Natural Resources, University of Missouri, Columbia, MO, U.S.A
| | - Mohamed Bayati
- Center for Agroforestry, University of Missouri, Columbia, MO, U.S.A
- School of Natural Resources, University of Missouri, Columbia, MO, U.S.A
- Environmental Engineering Department, Tikrit University, Tikrit, Iraq
| | - Novianus Efrat
- Center for Agroforestry, University of Missouri, Columbia, MO, U.S.A
- School of Natural Resources, University of Missouri, Columbia, MO, U.S.A
| | - Anuradha Roy
- High Throughput Screening Laboratory, University of Kansas, Lawrence, KS
| | - Zhentian Lei
- Division of Biochemistry, Bond Life Sciences Center, Interdisciplinary Plant Group, MU Metabolomics Center
| | - Khanh-Van Ho
- Division of Biochemistry, Bond Life Sciences Center, Interdisciplinary Plant Group, MU Metabolomics Center
| | - Lloyd Sumner
- Division of Biochemistry, Bond Life Sciences Center, Interdisciplinary Plant Group, MU Metabolomics Center
| | - Michael Greenlief
- Department of Chemistry, University of Missouri, Columbia, MO, U.S.A
| | - Andrew L. Thomas
- Division of Plant Science and Technology, Southwest Research, Extension, and Education Center, University of Missouri, Mt. Vernon, MO, U.S.A
| | | | | | - Chung-Ho Lin
- Center for Agroforestry, University of Missouri, Columbia, MO, U.S.A
- School of Natural Resources, University of Missouri, Columbia, MO, U.S.A
| |
Collapse
|
38
|
Liu L, He G, Li Y, Xian Y, He G, Hong Y, Zhang C, Wu D. Hyaluronic Acid-Based Microparticles with Lubrication and Anti-Inflammation for Alleviating Temporomandibular Joint Osteoarthritis. Biomater Res 2024; 28:0073. [PMID: 39247653 PMCID: PMC11377958 DOI: 10.34133/bmr.0073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/10/2024] [Indexed: 09/10/2024] Open
Abstract
The pathogenesis of temporomandibular joint osteoarthritis (TMJOA) is closely associated with mechanical friction, which leads to the up-regulation of inflammatory mediators and the degradation of articular cartilage. Injectable drug-loaded microparticles have attracted widespread interest in intra-articular treatment of TMJOA by providing lubrication and facilitating localized drug delivery. Herein, a hyaluronic acid-based microparticle is developed with excellent lubrication properties, drug loading capacity, antioxidant activity, and anti-inflammatory effect for the treatment of TMJOA. The microparticles are facilely prepared by the self-assembly of 3-aminophenylboronic acid-modified hyaluronic acid (HP) through hydrophobic interaction in an aqueous solution, which can further encapsulate diol-containing drugs through dynamic boronate ester bonds. The resulting microparticles demonstrate excellent injectability, lubrication properties, radical scavenging efficiency, and antibacterial activity. Additionally, the drug-loaded microparticles exhibit a favorable cytoprotective effect on chondrocyte cells in vitro under an oxidative stress microenvironment. In vivo experiments validate that intra-articular injection of drug-loaded microparticles effectively alleviates osteoporosis-like damage, suppresses inflammatory response, and facilitates matrix regeneration in the treatment of TMJOA. The HP microparticles demonstrate excellent injectability and encapsulation capacity for diol-containing drugs, highlighting its potential as a versatile drug delivery vehicle in the intra-articular treatment of TMJOA.
Collapse
Affiliation(s)
- Lei Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Gang He
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yixi Li
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yiwen Xian
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guixian He
- Department of Maxillofacial Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Yonglong Hong
- Department of Maxillofacial Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Chong Zhang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Decheng Wu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
39
|
Talib WH, Baban MM, Bulbul MF, Al-Zaidaneen E, Allan A, Al-Rousan EW, Ahmad RHY, Alshaeri HK, Alasmari MM, Law D. Natural Products and Altered Metabolism in Cancer: Therapeutic Targets and Mechanisms of Action. Int J Mol Sci 2024; 25:9593. [PMID: 39273552 PMCID: PMC11394730 DOI: 10.3390/ijms25179593] [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: 06/13/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Cancer is characterized by uncontrolled cell proliferation and the dysregulation of numerous biological functions, including metabolism. Because of the potential implications of targeted therapies, the metabolic alterations seen in cancer cells, such as the Warburg effect and disruptions in lipid and amino acid metabolism, have gained attention in cancer research. In this review, we delve into recent research examining the influence of natural products on altered cancer metabolism. Natural products were selected based on their ability to target cancer's altered metabolism. We identified the targets and explored the mechanisms of action of these natural products in influencing cellular energetics. Studies discussed in this review provide a solid ground for researchers to consider natural products in cancer treatment alone and in combination with conventional anticancer therapies.
Collapse
Affiliation(s)
- Wamidh H Talib
- Faculty of Allied Medical Sciences, Applied Science Private University, Amman 11931, Jordan
- Faculty of Health and Life Sciences, Inti International University, Nilai 71800, Negeri Sembilan, Malaysia
| | - Media Mohammad Baban
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan
| | - Mais Fuad Bulbul
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan
| | - Esraa Al-Zaidaneen
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan
| | - Aya Allan
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan
| | - Eiman Wasef Al-Rousan
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan
| | - Rahaf Hamed Yousef Ahmad
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan
| | - Heba K Alshaeri
- Department of Pharmacology, Faculty of Medicine, King Abdul-Aziz University, Rabigh 25724, Saudi Arabia
| | - Moudi M Alasmari
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah 21423, Saudi Arabia
- King Abdullah International Medical Research Centre (KAIMRC), Jeddah 22233, Saudi Arabia
| | - Douglas Law
- Faculty of Health and Life Sciences, Inti International University, Nilai 71800, Negeri Sembilan, Malaysia
| |
Collapse
|
40
|
Gu X, Wei S, Lv X. Circulating tumor cells: from new biological insights to clinical practice. Signal Transduct Target Ther 2024; 9:226. [PMID: 39218931 PMCID: PMC11366768 DOI: 10.1038/s41392-024-01938-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The primary reason for high mortality rates among cancer patients is metastasis, where tumor cells migrate through the bloodstream from the original site to other parts of the body. Recent advancements in technology have significantly enhanced our comprehension of the mechanisms behind the bloodborne spread of circulating tumor cells (CTCs). One critical process, DNA methylation, regulates gene expression and chromosome stability, thus maintaining dynamic equilibrium in the body. Global hypomethylation and locus-specific hypermethylation are examples of changes in DNA methylation patterns that are pivotal to carcinogenesis. This comprehensive review first provides an overview of the various processes that contribute to the formation of CTCs, including epithelial-mesenchymal transition (EMT), immune surveillance, and colonization. We then conduct an in-depth analysis of how modifications in DNA methylation within CTCs impact each of these critical stages during CTC dissemination. Furthermore, we explored potential clinical implications of changes in DNA methylation in CTCs for patients with cancer. By understanding these epigenetic modifications, we can gain insights into the metastatic process and identify new biomarkers for early detection, prognosis, and targeted therapies. This review aims to bridge the gap between basic research and clinical application, highlighting the significance of DNA methylation in the context of cancer metastasis and offering new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Xuyu Gu
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
41
|
Geng Q, Yan L, Shi C, Zhang L, Li L, Lu P, Cao Z, Li L, He X, Tan Y, Zhao N, Liu B, Lu C. Therapeutic effects of flavonoids on pulmonary fibrosis: A preclinical meta-analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155807. [PMID: 38876010 DOI: 10.1016/j.phymed.2024.155807] [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: 04/01/2024] [Revised: 05/26/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND The efficacy of flavonoid supplementation in animal models of pulmonary fibrosis has been demonstrated. PURPOSE We conducted a systematic review and meta-analysis to evaluate the efficacy and underlying mechanisms of flavonoids in animal models of bleomycin-induced pulmonary fibrosis. STUDY DESIGN Relevant studies (n = 45) were identified from English- and Chinese-language databases from the inception of the database until October 2023. METHODS Methodological quality was evaluated using the SYRCLE risk of bias tool. Statistical analyses were conducted using RevMan 5.3 and Stata 17.0. Lung inflammation and fibrosis score were the primary outcome indicators. RESULTS Flavonoids can alleviate pathological changes in the lungs. The beneficial effects of flavonoids on pulmonary fibrosis likely relate to their inhibition of inflammatory responses, restoration of oxidative and antioxidant homeostasis, and regulation of fibroblast proliferation, migration, and activation by transforming growth factor β1/mothers against the decapentaplegic homologue/AMP-activated protein kinase (TGF-β1/Smad3/AMPK), inhibitor kappa B alpha/nuclear factor-kappa B (IκBα/NF-κB), phosphatidylinositol 3-kinase (PI3K)/AKT, interleukin 6/signal transducer/activator of transcription 3 (IL6/STAT3), and nuclear factor erythroid 2-related factor 2/Kelch-like ECH-associated protein 1 (Nrf2-Keap1) pathways. CONCLUSION Flavonoids are potential candidate compounds for the prevention and treatment of pulmonary fibrosis. However, extensive preclinical research is necessary to confirm the antifibrotic properties of natural flavonoids.
Collapse
Affiliation(s)
- Qi Geng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Lan Yan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Changqi Shi
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Lulu Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Peipei Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Zhiwen Cao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Yong Tan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Bin Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China.
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, PR China.
| |
Collapse
|
42
|
Ye XS, Lin K, Tao XQ, Shang JT, Gui YR, Zhu SX, Zhou L, Xia YY, Liu W, Sun BL, Chen HF, Shu XJ. Discovery of Sesquiterpene Lactones with Cytotoxicity from the Herb of Youngia japonica. Chem Biodivers 2024; 21:e202401063. [PMID: 38924351 DOI: 10.1002/cbdv.202401063] [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: 04/26/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
In the process of searching for anti-breast cancer agents, five sesquiterpene lactones (1-5), including two previously undescribed ones, yjaponica B-C (1-2), were isolated from the herb of Youngia japonica. Their structures were elucidated by spectroscopic data analyses and Marfey's method. Cytotoxic activities of all compounds against A549, U87, and 4T1 cell lines were tested using the CCK8 assay. The result showed that compound 3 possessed the highest cytotoxic activity against 4T1 cells with an IC50 value of 10.60 μM. Furthermore, compound 3 distinctly induced apoptosis, inhibited immigration, and blocked the cell cycle of 4T1 cells. In addition, compound 3 induced the production of reactive oxygen species. Further anticancer mechanism studies showed that compound 3 significantly upregulated expression of the cleaved caspase 3 and PARP, whereas it downregulated the expression of Bcl-2, cyclin D1, cyclin A2, CDK4, and CDK2. Taken together, our results demonstrate that compound 3 has a high potential of being used as a leading compound for the discovery of new anti-breast cancer agent.
Collapse
Affiliation(s)
- Xian-Sheng Ye
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Kuan Lin
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Xiao-Qing Tao
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Jin-Ting Shang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Yu-Ran Gui
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Shu-Xiu Zhu
- Department of Traditional Chinese Medicine, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Le Zhou
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, RNAM Center for Marine Microbiology, Chinese Academy of Sciences, South China Sea Institute of Oceanology, Guangzhou, 510000, China
| | - Yi-Yuan Xia
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Wei Liu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Bin-Lian Sun
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Hai-Feng Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Xi-Ji Shu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| |
Collapse
|
43
|
Sun Z, Wei Y, Xu Y, Jiao J, Duan X. The use of traditional Chinese medicine in the treatment of non-alcoholic fatty liver disease: A review. PHARMACOLOGICAL RESEARCH - MODERN CHINESE MEDICINE 2024; 12:100475. [DOI: 10.1016/j.prmcm.2024.100475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
44
|
Li M, Wang H, Bai Y, Xiong F, Wu S, Bi Q, Qiao Y, Zhang Y, Li X, Feng L, Guo DA. Pharmacodynamical research of extracts and compounds in traditional Chinese medicines for Parkinson's disease. Fitoterapia 2024; 177:106086. [PMID: 38897243 DOI: 10.1016/j.fitote.2024.106086] [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: 05/12/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease (AD). Currently, there is no cure for PD, and medications can only control the progression of the disease. Various experimental studies have shown the significant efficacy of TCM in treating PD, and combination with western medicine can enhance the effects and reduce toxicity. Thus, exploring effective anti-PD compounds from TCM has become a popular research fields. This review summarizes commonly used TCM extracts and natural products for the treatment of PD, both domestically and internationally. Furthermore, it delves into various mechanisms of TCM in treating PD, such as anti-oxidative stress, anti-inflammatory, anti-apoptotic, improve mitochondrial dysfunction, inhibits α-synuclein (α-Syn) misfolding and aggregation, regulating neurotransmitters, regulates intestinal flora, enhances immunity, and so on. The results reveal that most TCMs exert their neuroprotective effects through anti-inflammatory and anti-oxidative stress actions, thereby slowing down the progression of the disease. These TCM may hold the key to improving PD therapy and have tremendous potential to be developed as novel anti-PD drugs.
Collapse
Affiliation(s)
- Mengmeng Li
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hanze Wang
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuxin Bai
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fuyu Xiong
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shifei Wu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qirui Bi
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yajun Qiao
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yan Zhang
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaolan Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lin Feng
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - De-An Guo
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
45
|
Cai S, Gou Y, Chen Y, Hou X, Zhang J, Bi C, Gu P, Yang M, Zhang H, Zhong W, Yuan H. Luteolin exerts anti-tumour immunity in hepatocellular carcinoma by accelerating CD8 + T lymphocyte infiltration. J Cell Mol Med 2024; 28:e18535. [PMID: 39267250 PMCID: PMC11392827 DOI: 10.1111/jcmm.18535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 09/17/2024] Open
Abstract
Luteolin, a commonly used traditional Chinese medicine, has been utilized for several decades in the treatment of hepatocellular carcinoma (HCC). Previous research has demonstrated its anti-tumour efficacy, but its underlying mechanism remains unclear. This study aimed to assess the therapeutic effects of luteolin in H22 tumour-bearing mice. luteolin effectively inhibited the growth of solid tumours in a well-established mouse model of HCC. High-throughput sequencing revealed that luteolin treatment could enhance T-cell activation, cell chemotaxis and cytokine production. In addition, luteolin helped sustain a high ratio of CD8+ T lymphocytes in the spleen, peripheral blood and tumour tissues. The effects of luteolin on the phenotypic and functional changes in tumour-infiltrating CD8+ T lymphocytes were also investigated. Luteolin restored the cytotoxicity of tumour-infiltrating CD8+ T lymphocytes in H22 tumour-bearing mice. The CD8+ T lymphocytes exhibited intensified phenotype activation and increased production of granzyme B, IFN-γ and TNF-α in serum. The combined administration of luteolin and the PD-1 inhibitor enhanced the anti-tumour effects in H22 tumour-bearing mice. Luteolin could exert an anti-tumour immune response by inducing CD8+ T lymphocyte infiltration and enhance the anti-tumour effects of the PD-1 inhibitor on H22 tumour-bearing mice.
Collapse
Affiliation(s)
- Shijiao Cai
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Yidan Gou
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Yanyan Chen
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoran Hou
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Zhang
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Chongwen Bi
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Gu
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Miao Yang
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Hanxu Zhang
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Weilong Zhong
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Hengjie Yuan
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
46
|
Al Azzani M, Nizami ZN, Magramane R, Sekkal MN, Eid AH, Al Dhaheri Y, Iratni R. Phytochemical-mediated modulation of autophagy and endoplasmic reticulum stress as a cancer therapeutic approach. Phytother Res 2024; 38:4353-4385. [PMID: 38961675 DOI: 10.1002/ptr.8283] [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: 01/20/2024] [Revised: 05/30/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
Autophagy and endoplasmic reticulum (ER) stress are conserved processes that generally promote survival, but can induce cell death when physiological thresholds are crossed. The pro-survival aspects of these processes are exploited by cancer cells for tumor development and progression. Therefore, anticancer drugs targeting autophagy or ER stress to induce cell death and/or block the pro-survival aspects are being investigated extensively. Consistently, several phytochemicals have been reported to exert their anticancer effects by modulating autophagy and/or ER stress. Various phytochemicals (e.g., celastrol, curcumin, emodin, resveratrol, among others) activate the unfolded protein response to induce ER stress-mediated apoptosis through different pathways. Similarly, various phytochemicals induce autophagy through different mechanisms (namely mechanistic target of Rapamycin [mTOR] inhibition). However, phytochemical-induced autophagy can function either as a cytoprotective mechanism or as programmed cell death type II. Interestingly, at times, the same phytochemical (e.g., 6-gingerol, emodin, shikonin, among others) can induce cytoprotective autophagy or programmed cell death type II depending on cellular contexts, such as cancer type. Although there is well-documented mechanistic interplay between autophagy and ER stress, only a one-way modulation was noted with some phytochemicals (carnosol, capsaicin, cryptotanshinone, guangsangon E, kaempferol, and δ-tocotrienol): ER stress-dependent autophagy. Plant extracts are sources of potent phytochemicals and while numerous phytochemicals have been investigated in preclinical and clinical studies, the search for novel phytochemicals with anticancer effects is ongoing from plant extracts used in traditional medicine (e.g., Origanum majorana). Nonetheless, the clinical translation of phytochemicals, a promising avenue for cancer therapeutics, is hindered by several limitations that need to be addressed in future studies.
Collapse
Affiliation(s)
- Mazoun Al Azzani
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rym Magramane
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed N Sekkal
- Department of Surgery, Specialty Orthopedic, Tawam Hospital, Al Ain, United Arab Emirates
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Yusra Al Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
47
|
Wang R, Li X, Xu Y, Li Y, Zhang W, Guo R, Song J. Progress, pharmacokinetics and future perspectives of luteolin modulating signaling pathways to exert anticancer effects: A review. Medicine (Baltimore) 2024; 103:e39398. [PMID: 39183411 PMCID: PMC11346905 DOI: 10.1097/md.0000000000039398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024] Open
Abstract
Luteolin (3, 4, 5, 7-tetrahydroxyflavone) are natural flavonoids widely found in vegetables, fruits and herbs, with anti-tumor, anti-inflammatory and antioxidant effects, and also play an anti-cancer effect in various cancers such as lung, breast, prostate, and liver cancer, etc. Specifically, the anti-cancer mechanism includes regulation of various signaling pathways to induce apoptosis of tumor cells, inhibition of tumor cell proliferation and metastasis, anti-angiogenesis, regulation of immune function, synergistic anti-cancer drugs and regulation of reactive oxygen species levels of tumor cells. Specific anti-cancer mechanisms include regulation of various signaling pathways to induce apoptosis, inhibition of tumor cell proliferation and metastasis, anti-angiogenesis, reversal of epithelial-mesenchymal transition, regulation of immune function, synergism with anti-cancer drugs and regulation of reactive oxygen species levels in tumor cells. This paper integrates the latest cutting-edge research on luteolin and combines it with the prospect of future clinical applications, aiming to explore the mechanism of luteolin exerting different anticancer effects through the regulation of different signaling pathways, so as to provide a practical theoretical basis for the use of luteolin in clinical treatment and hopefully provide some reference for the future research direction of luteolin.
Collapse
Affiliation(s)
- Rui Wang
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
- Medical School of Nantong University, Nantong, PR China
| | - Xia Li
- Department of General Medicine, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Yanhan Xu
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Yangyang Li
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Weisong Zhang
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Rongqi Guo
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Jianxiang Song
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| |
Collapse
|
48
|
Valivand N, Aravand S, Lotfi H, Esfahani AJ, Ahmadpour-Yazdi H, Gheibi N. Propolis: a natural compound with potential as an adjuvant in cancer therapy - a review of signaling pathways. Mol Biol Rep 2024; 51:931. [PMID: 39177837 DOI: 10.1007/s11033-024-09807-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 07/18/2024] [Indexed: 08/24/2024]
Abstract
Propolis is a natural product used in cancer treatment, which is produced by bees via different sources. The chemical composition of Propolis is determined based on the climatic and geographical conditions, as well as harvesting time and method. This compound has been the subject of numerous investigational endeavors due to its expansive therapeutic capacity which includes antibacterial, anti-fungal, anti-inflammatory, anti-oxidant, anti-viral, and anti-cancer effects. The growing incidence rate of different cancers necessitates the need for developing novel preventive and therapeutic strategies. Chemotherapy, radiotherapy, and stem cell therapy have proved effective in cancer treatment, regardless of the adverse events associated with these modalities. Clinical application of natural compounds such as Propolis may confer promise as an adjuvant therapeutic intervention, particularly in certain subpopulations of patients that develop adverse events associated with anticancer regimens. The diverse biologically active compounds of propolis are believed to confer anti-cancer potential by modulation of critical signaling cascades such as caffeic acid phenethyl ester, Galangin, Artepillin C, Chrysin, Quercetin, Caffeic acid, Nymphaeols A and C, Frondoside A, Genistein, p-coumaric acid, and Propolin C. This review article aims to deliver a mechanistic account of anti-cancer effects of propolis and its components. Propolis can prevent angiogenesis by downregulating pathways involving Jun-N terminal kinase, ERK1/2, Akt and NF-ƘB, while counteracting metastatic progression of cancer by inhibiting Wtn2 and FAK, and MAPK and PI3K/AKT signaling pathways. Moreover, propolis or its main components show regulatory effects on cyclin D, CDK2/4/6, and their inhibitors. Additionally, propolis-induced up-regulation of p21 and p27 may result in cell cycle arrest at G2/M or G0/G1. The broad anti-apoptotic effects of propolis are mediated through upregulation of TRAIL, Bax, p53, and downregulation of the ERK1/2 signaling pathway. Considering the growing body of evidence regarding different anti-cancers effects of propolis and its active components, this natural compound could be considered an effective adjuvant therapy aimed at reducing related side effects associated with chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Nassim Valivand
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sara Aravand
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hajie Lotfi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Azam Janati Esfahani
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hossein Ahmadpour-Yazdi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Nematollah Gheibi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
49
|
Ma J, Mo J, Feng Y, Wang L, Jiang H, Li J, Jin C. Combination of transcriptomic and proteomic approaches helps unravel the mechanisms of luteolin in inducing liver cancer cell death via targeting AKT1 and SRC. Front Pharmacol 2024; 15:1450847. [PMID: 39234106 PMCID: PMC11371790 DOI: 10.3389/fphar.2024.1450847] [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: 06/18/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Luteolin, a natural compound commonly used in traditional Chinese medicine, shows clinical potential as an anti-liver cancer agent. The mechanisms underlying the anti-liver cancer effect of luteolin are limited versus those reported for other cancers. Accordingly, this study was conducted to bridge the existing knowledge gap. Methods Transcriptomic and proteomic analyses of the response of the hepatocellular carcinoma cell line HuH-7 to luteolin were conducted, and a possible pathway was elucidated using confocal laser scanning microscopy (CLSM), flow cytometry, western blotting, qRT-PCR and bio-layer interferometry assay to systematically explore the possible mechanisms underlying the inhibition of the proliferation of liver cancer cells by luteolin. Results and Discussion Results showed that luteolin significantly inhibited HuH-7 cell proliferation. Transcriptomic and proteomic analyses collectively revealed that luteolin could promote cell cycle arrest and apoptosis in HuH-7 cells through transcription factors p53, nuclear factor kappa B (NF-κB), FOXO, ATF2, and TCF/LEF via AKT1, as well as the KEAP-NRF and SRC-STAT3 pathways. Furthermore, AKT1 and SRC were identified as the 2 targets of luteolin. Nuclear translocation of transcription factors p53 and NF-κB were affected by luteolin administration. Additionally, AKT1 activity affected normal metabolism in HuH-7 cells and resulted in the accumulation of reactive oxygen species, which activated MOMP and further promoted apoptosis. Our results systematically elucidate the mechanism of luteolin in inhibiting the proliferation of liver cancer cells, mainly through cell cycle arrest and apoptosis via targeting AKT1 and SRC.
Collapse
Affiliation(s)
- Junxia Ma
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Evolutionary Ecology and Conservation, Taizhou University, Taizhou, China
| | - Jinggang Mo
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Yifu Feng
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Liezhi Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Hao Jiang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Junmin Li
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Evolutionary Ecology and Conservation, Taizhou University, Taizhou, China
| | - Chong Jin
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
50
|
Móritz AV, Kovács H, Jerzsele Á, Psáder R, Farkas O. Flavonoids in mitigating the adverse effects of canine endotoxemia. Front Vet Sci 2024; 11:1396870. [PMID: 39193369 PMCID: PMC11347451 DOI: 10.3389/fvets.2024.1396870] [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: 03/06/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
In dogs, chronic enteropathies, and impaired gut integrity, as well as microbiome imbalances, are a major problem. These conditions may represent a continuous low endotoxin load, which may result in the development of diseases that are attributable to chronic inflammation. Flavonoids are polyphenolic plant compounds with numerous beneficial properties such as antioxidant, anti-inflammatory and antimicrobial effects. For our experiments, we isolated primary white blood cells (peripheral blood mononuclear cells and polymorphonuclear leukocytes) from healthy dogs and induced inflammation and oxidative stress with Escherichia coli and Salmonella enterica serovar Enteritidis lipopolysaccharide (LPS). In parallel, we treated the cell cultures with various flavonoids luteolin, quercetin and grape seed extract oligomeric proanthocyanidins (GSOP) alone and also in combination with LPS treatments. Then, changes in viability, reactive oxygen species (ROS) and tumor necrosis factor alpha (TNF-α) levels were measured in response to treatment with quercetin, luteolin and GSOP at 25 and 50 μg/mL concentrations. We found that ROS levels were significantly lower in groups which were treated by flavonoid and LPS at the same time compared to LPS-treated groups, whereas TNF-α levels were significantly reduced only by luteolin and quercetin treatment. In contrast, treatment with lower concentrations of GSOP caused an increase in TNF-α levels, while higher concentrations caused a significant decrease. These results suggest that the use of quercetin, luteolin and GSOP may be helpful in the management of chronic intestinal diseases in dogs with reduced intestinal barrier integrity or altered microbiome composition, or in the mitigation of chronic inflammatory processes maintained by endotoxemia. Further in vitro and in vivo studies are needed before clinical use.
Collapse
Affiliation(s)
- Alma V. Móritz
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, Hungary
| | - Hédi Kovács
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, Hungary
| | - Roland Psáder
- Department of Internal Medicine, University of Veterinary Medicine, Budapest, Hungary
| | - Orsolya Farkas
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, Hungary
| |
Collapse
|