1
|
Su L, Bu J, Yu J, Jin M, Meng G, Zhu X. Comprehensive review and updated analysis of DNA methylation in hepatocellular carcinoma: From basic research to clinical application. Clin Transl Med 2024; 14:e70066. [PMID: 39462685 PMCID: PMC11513202 DOI: 10.1002/ctm2.70066] [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: 06/04/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary malignant tumour, ranking second in global mortality rates and posing significant health threats. Epigenetic alterations, particularly DNA methylation, have emerged as pivotal factors associated with HCC diagnosis, therapy, prognosis and malignant progression. However, a comprehensive analysis of the DNA methylation mechanism driving HCC progression and its potential as a therapeutic biomarker remains lacking. This review attempts to comprehensively summarise various aspects of DNA methylation, such as its mechanism, detection methods and biomarkers aiding in HCC diagnosis, treatment and prognostic assessment of HCC. It also explores the role of DNA methylation in regulating HCC's malignant progression and sorafenib resistance, alongside elaborating the therapeutic effects of DNA methyltransferase inhibitors on HCC. A detailed examination of these aspects underscores the significant research on DNA methylation in tumour cells to elucidate malignant progression mechanisms, identify diagnostic markers and develop new tumour-specific inhibitors for HCC. KEY POINTS: A comprehensive summary of various aspects of DNA methylation, such as its mechanism, detection methods and biomarkers aiding in diagnosis and treatment. The role of DNA methylation in regulating hepatocellular carcinoma's (HCC) malignant progression and sorafenib resistance, alongside elaborating therapeutic effects of DNA methyltransferase inhibitors. Deep research on DNA methylation is critical for discovering novel tumour-specific inhibitors for HCC.
Collapse
Affiliation(s)
- Lin Su
- Department of Pain ManagementShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jiawen Bu
- Department of Colorectal SurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jiahui Yu
- Department of UltrasoundShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Mila Jin
- Department of Operation RoomThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Guanliang Meng
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xudong Zhu
- Department of OncologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Department of General SurgeryCancer Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
2
|
Bouyahya A, Bakrim S, Aboulaghras S, El Kadri K, Aanniz T, Khalid A, Abdalla AN, Abdallah AA, Ardianto C, Ming LC, El Omari N. Bioactive compounds from nature: Antioxidants targeting cellular transformation in response to epigenetic perturbations induced by oxidative stress. Biomed Pharmacother 2024; 174:116432. [PMID: 38520868 DOI: 10.1016/j.biopha.2024.116432] [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: 12/17/2023] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024] Open
Abstract
Oxidative stress results from a persistent imbalance in oxidation levels that promotes oxidants, playing a crucial role in the early and sustained phases of DNA damage and genomic and epigenetic instability, both of which are intricately linked to the development of tumors. The molecular pathways contributing to carcinogenesis in this context, particularly those related to double-strand and single-strand breaks in DNA, serve as indicators of DNA damage due to oxidation in cancer cases, as well as factors contributing to epigenetic instability through ectopic expressions. Oxidative stress has been considered a therapeutic target for many years, and an increasing number of studies have highlighted the promising effectiveness of natural products in cancer treatment. In this regard, we present significant research on the therapeutic targeting of oxidative stress using natural molecules and underscore the essential role of oxidative stress in cancer. The consequences of stress, especially epigenetic instability, also offer significant therapeutic prospects. In this context, the use of natural epi-drugs capable of modulating and reorganizing the epigenetic network is beginning to emerge remarkably. In this review, we emphasize the close connections between oxidative stress, epigenetic instability, and tumor transformation, while highlighting the role of natural substances as antioxidants and epi-drugs in the anti-tumoral context.
Collapse
Affiliation(s)
- Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco.
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Sara Aboulaghras
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Kawtar El Kadri
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Tarik Aanniz
- Biotechnology Lab (MedBiotech), Bioinova Research Center, Rabat Medical & Pharmacy School, Mohammed V University in Rabat, Morocco
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan PO Box: 114, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ahmed A Abdallah
- Department of Anatomy, Faculty of Medicine, Umm Alqura University, Makkah 21955, Saudi Arabia
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia.
| | - Long Chiau Ming
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia; School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia; Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam.
| | - Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| |
Collapse
|
3
|
Yang F, Yang Y, Qiu Y, Tang L, Xie L, Guan X. Long Non-Coding RNAs as Regulators for Targeting Breast Cancer Stem Cells and Tumor Immune Microenvironment: Biological Properties and Therapeutic Potential. Cancers (Basel) 2024; 16:290. [PMID: 38254782 PMCID: PMC10814583 DOI: 10.3390/cancers16020290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/01/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Breast cancer stem cells (BCSCs) is a subpopulation of cancer cells with self-renewal and differentiation capacity, have been suggested to give rise to tumor heterogeneity and biologically aggressive behavior. Accumulating evidence has shown that BCSCs play a fundamental role in tumorigenesis, progression, and recurrence. The development of immunotherapy, primarily represented by programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) inhibitors, has greatly changed the treatment landscape of multiple malignancies. Recent studies have identified pervasive negative associations between cancer stemness and anticancer immunity. Stemness seems to play a causative role in the formation of cold tumor immune microenvironment (TIME). The multiple functions of long non-coding RNAs (lncRNAs) in regulating stemness and immune responses has been recently highlighted in breast cancer. The review focus on lncRNAs and keys pathways involved in the regulation of BCSCs and TIME. Potential clinical applications using lncRNAs as biomarkers or therapies will be discussed.
Collapse
Affiliation(s)
- Fang Yang
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; (F.Y.); (Y.Y.); (Y.Q.)
- Clinical Cancer Institute, Nanjing University, Nanjing 210008, China
| | - Yiqi Yang
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; (F.Y.); (Y.Y.); (Y.Q.)
- Clinical Cancer Institute, Nanjing University, Nanjing 210008, China
| | - Yuling Qiu
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; (F.Y.); (Y.Y.); (Y.Q.)
- Clinical Cancer Institute, Nanjing University, Nanjing 210008, China
| | - Lin Tang
- Department of Rheumatology and Immunology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China;
| | - Li Xie
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; (F.Y.); (Y.Y.); (Y.Q.)
- Clinical Cancer Institute, Nanjing University, Nanjing 210008, China
| | - Xiaoxiang Guan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
4
|
Qadir Nanakali NM, Maleki Dana P, Sadoughi F, Asemi Z, Sharifi M, Asemi R, Yousefi B. The role of dietary polyphenols in alternating DNA methylation in cancer. Crit Rev Food Sci Nutr 2023; 63:12256-12269. [PMID: 35848113 DOI: 10.1080/10408398.2022.2100313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Natural products such as curcumin, quercetin, and resveratrol have been shown to have antitumor effectsand several studies have examined their role in treating cancer, either alone or in combination with other chemotherapeutic drugs. These compounds are capable of affecting different cancer-related mechanisms, such as proliferation, inflammation, invasion, and metastasis. Along with all of the benefits of these agents, affecting epigenetic processes is one of the most important aspects of their impact. Epigenetic modifications can be categorized into three main processes that include DNA methylation, histone modification, and regulation of small non-coding RNAs. Therefore, targeting DNA methylation can be used as a cancer treatment strategy by identifying or developing methylation modulators. Herein, we take a look into the studies investigating the role of natural products (e.g. curcumin, resveratrol, epigallocatechin gallate (EGCG), and quercetin) in alternating the DNA methylation status of various cancer cells. We discuss how these compounds reduce the expression of enzymes mediating the methylation of tumor suppressor genes and thereby, increasing the expression of tumor suppressors while reactivating antitumor signaling pathways.
Collapse
Affiliation(s)
- Nadir Mustafa Qadir Nanakali
- Department of Biomedical Science, College of Science, Cihan University-Erbil, Kurdistan Region, Erbil, Iraq
- Department of Biology, College of Education, Salahaddin University-Erbil, Kurdistan Region, Erbil, Iraq
| | - Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Mehran Sharifi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Asemi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
Ordaz-Ramos A, Tellez-Jimenez O, Vazquez-Santillan K. Signaling pathways governing the maintenance of breast cancer stem cells and their therapeutic implications. Front Cell Dev Biol 2023; 11:1221175. [PMID: 37492224 PMCID: PMC10363614 DOI: 10.3389/fcell.2023.1221175] [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: 05/11/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023] Open
Abstract
Breast cancer stem cells (BCSCs) represent a distinct subpopulation of cells with the ability to self-renewal and differentiate into phenotypically diverse tumor cells. The involvement of CSC in treatment resistance and cancer recurrence has been well established. Numerous studies have provided compelling evidence that the self-renewal ability of cancer stem cells is tightly regulated by specific signaling pathways, which exert critical roles to maintain an undifferentiated phenotype and prevent the differentiation of CSCs. Signaling pathways such as Wnt/β-catenin, NF-κB, Notch, Hedgehog, TGF-β, and Hippo have been implicated in the promotion of self-renewal of many normal and cancer stem cells. Given the pivotal role of BCSCs in driving breast cancer aggressiveness, targeting self-renewal signaling pathways holds promise as a viable therapeutic strategy for combating this disease. In this review, we will discuss the main signaling pathways involved in the maintenance of the self-renewal ability of BCSC, while also highlighting current strategies employed to disrupt the signaling molecules associated with stemness.
Collapse
Affiliation(s)
- Alejandro Ordaz-Ramos
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, México
| | - Olivia Tellez-Jimenez
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, México
| | - Karla Vazquez-Santillan
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
| |
Collapse
|
6
|
Yang Y, Jin M, Meng Y, Dai Y, Chen S, Zhou Y, Li Y, Tang L. Involvement and targeted intervention of benzo(a)pyrene-regulated apoptosis related proteome modification and muti-drug resistance in hepatocellular carcinoma. Cell Death Dis 2023; 14:265. [PMID: 37041133 PMCID: PMC10090052 DOI: 10.1038/s41419-023-05771-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/13/2023]
Abstract
During the development of hepatocellular carcinoma (HCC), the mutual adaptation and interaction of HCC cells and the microenvironment play an important role. Benzo(a)pyrene (B[a]P) is a common environmental pollutant, which can induce the initiation of various malignant tumors, including HCC. However, the effects of B[a]P exposure on progression of HCC and the potential mechanisms remains largely uninvestigated. Here we found that, after the long-term exposure of HCC cells to low dose of B[a]P, it activated glucose-regulated protein 75 (GRP75), which then induced a modification of apoptosis-related proteome. Among them, we identified the X-linked inhibitor of apoptosis protein (XIAP) as a key downstream factor. XIAP further blocked the caspase cascade activation and promoted the acquisition of the anti-apoptosis abilities, ultimately leading to multi-drug resistance (MDR) in HCC. Furthermore, the abovementioned effects were markedly attenuated when we inhibited GRP75 by using 3,4-dihydroxycinnamic acid (caffeic acid, CaA). Collectively, our present study revealed the effects of B[a]P exposure on the progression of HCC, and identified GRP75 was a meaningful factor involved in.
Collapse
Affiliation(s)
- Ye Yang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ming Jin
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yajie Meng
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yi Dai
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Shuai Chen
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Yan Zhou
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Yuan Li
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213003, China.
| | - Liming Tang
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213003, China.
| |
Collapse
|
7
|
Ding Q, Xu YM, Lau ATY. The Epigenetic Effects of Coffee. Molecules 2023; 28:molecules28041770. [PMID: 36838754 PMCID: PMC9958838 DOI: 10.3390/molecules28041770] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 02/16/2023] Open
Abstract
In this review, we discuss the recent knowledge regarding the epigenetic effects of coffee extract and the three essential active ingredients in coffee (caffeine, chlorogenic acid, and caffeic acid). As a popular beverage, coffee has many active ingredients which have a variety of biological functions such as insulin sensitization, improvement of sugar metabolism, antidiabetic properties, and liver protection. However, recent researches have shown that coffee is not only beneficial for human, but also bad, which may be due to its complex components. Studies suggest that coffee extract and its components can potentially impact gene expression via alteration of DNA methylation, histone modifications, and ncRNA expression; thus, exert long lasting impacts on the epigenome. More importantly, coffee consumption during pregnancy has been linked to multiple negative effects on offspring due to epigenetic modifications; on the other hand, it has also been linked to improvements in many diseases, including cancer. Therefore, understanding more about the epigenetic effects associated with coffee components is crucial to finding ways for improving human health.
Collapse
Affiliation(s)
| | - Yan-Ming Xu
- Correspondence: (Y.-M.X.); (A.T.Y.L.); Tel.: +86-754-8890-0437 (Y.-M.X.); +86-754-8853-0052 (A.T.Y.L.)
| | - Andy T. Y. Lau
- Correspondence: (Y.-M.X.); (A.T.Y.L.); Tel.: +86-754-8890-0437 (Y.-M.X.); +86-754-8853-0052 (A.T.Y.L.)
| |
Collapse
|
8
|
Nazam N, Jabir NR, Ahmad I, Alharthy SA, Khan MS, Ayub R, Tabrez S. Phenolic Acids-Mediated Regulation of Molecular Targets in Ovarian Cancer: Current Understanding and Future Perspectives. Pharmaceuticals (Basel) 2023; 16:274. [PMID: 37259418 PMCID: PMC9962268 DOI: 10.3390/ph16020274] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is a global health concern with a dynamic rise in occurrence and one of the leading causes of mortality worldwide. Among different types of cancer, ovarian cancer (OC) is the seventh most diagnosed malignant tumor, while among the gynecological malignancies, it ranks third after cervical and uterine cancer and sadly bears the highest mortality and worst prognosis. First-line treatments have included a variety of cytotoxic and synthetic chemotherapeutic medicines, but they have not been particularly effective in extending OC patients' lives and are associated with side effects, recurrence risk, and drug resistance. Hence, a shift from synthetic to phytochemical-based agents is gaining popularity, and researchers are looking into alternative, cost-effective, and safer chemotherapeutic strategies. Lately, studies on the effectiveness of phenolic acids in ovarian cancer have sparked the scientific community's interest because of their high bioavailability, safety profile, lesser side effects, and cost-effectiveness. Yet this is a road less explored and critically analyzed and lacks the credibility of the novel findings. Phenolic acids are a significant class of phytochemicals usually considered in the nonflavonoid category. The current review focused on the anticancer potential of phenolic acids with a special emphasis on chemoprevention and treatment of OC. We tried to summarize results from experimental, epidemiological, and clinical studies unraveling the benefits of various phenolic acids (hydroxybenzoic acid and hydroxycinnamic acid) in chemoprevention and as anticancer agents of clinical significance.
Collapse
Affiliation(s)
- Nazia Nazam
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida 201301, Uttar Pradesh, India
| | - Nasimudeen R. Jabir
- Department of Biochemistry, Centre for Research and Development, PRIST University, Vallam, Thanjavur 613403, Tamil Nadu, India
| | - Iftikhar Ahmad
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Saif A. Alharthy
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Rashid Ayub
- Technology and Innovation Unit, Department of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
9
|
Abd Elhamid MA, Mandour AES, Ismail TA, Al-Zohairy AM, Almowallad S, Alqahtani LS, Osman A. Powerful Antioxidants and Cytotoxic Activities of the Methanol Extracts from Eight Soybean Cultivars. Molecules 2022; 27:2895. [PMID: 35566246 PMCID: PMC9104066 DOI: 10.3390/molecules27092895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 12/01/2022] Open
Abstract
In the present study, the chemical composition and total phenolic (TPC) and total flavonoid contents (TFC) of eight soybean cultivars (Giza 21, Giza 22, Giza 35, Giza 111, Giza 82, Giza 83, Crawford, and Holliday) were estimated. Moreover, antioxidant activity and in vitro cytotoxic activities against HepG-2 and MCF-7 were evaluated. Giza 21, Giza 111, and Crawford cultivars recorded higher than 40% crude protein. The analysis revealed that TPC values in seed extracts ranged from 10.5 mg GAE/g extract in Giza 35 to 6.4 mg GAE/g extract in Giza 22. TFC varied from 1.20 mg QE/g extract in Giza 111 to 0.55 mg QE/g extract in Crawford. Giza 35 exhibited the highest content of genistein and daidzein and the highest free radical scavenging activity (61.833%). The results of the MTT assay demonstrated that the soybean methanolic extracts inhibited the proliferation of HepG-2 and MCF-7 cells in a dose-dependent manner. Giza 35 exhibited the highest cytotoxic activity. In conclusion, Giza 35 cultivar recorded the highest TPC and TFC values and antioxidant and cytotoxic activities. Therefore, this cultivar can be used as a source for the production of pharmaceutical and medicinal products rather than as a nutritional source of protein.
Collapse
Affiliation(s)
- Mohamed A. Abd Elhamid
- Genetics Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt; (M.A.A.E.); (A.E.S.M.); (T.A.I.); (A.M.A.-Z.)
| | - Abd Elrahman S. Mandour
- Genetics Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt; (M.A.A.E.); (A.E.S.M.); (T.A.I.); (A.M.A.-Z.)
| | - Tarek A. Ismail
- Genetics Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt; (M.A.A.E.); (A.E.S.M.); (T.A.I.); (A.M.A.-Z.)
| | - Ahmed M. Al-Zohairy
- Genetics Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt; (M.A.A.E.); (A.E.S.M.); (T.A.I.); (A.M.A.-Z.)
| | - Sanaa Almowallad
- Department of Biochemistry, Faculty of Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Leena S. Alqahtani
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah 23445, Saudi Arabia;
| | - Ali Osman
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| |
Collapse
|
10
|
Alam M, Ashraf GM, Sheikh K, Khan A, Ali S, Ansari MM, Adnan M, Pasupuleti VR, Hassan MI. Potential Therapeutic Implications of Caffeic Acid in Cancer Signaling: Past, Present, and Future. Front Pharmacol 2022; 13:845871. [PMID: 35355732 PMCID: PMC8959753 DOI: 10.3389/fphar.2022.845871] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 01/26/2022] [Indexed: 12/20/2022] Open
Abstract
Caffeic acid (CA) has been present in many herbs, vegetables, and fruits. CA is a bioactive compound and exhibits various health advantages that are linked with its anti-oxidant functions and implicated in the therapy and prevention of disease progression of inflammatory diseases and cancer. The anti-tumor action of CA is attributed to its pro-oxidant and anti-oxidant properties. CA’s mechanism of action involves preventing reactive oxygen species formation, diminishing the angiogenesis of cancer cells, enhancing the tumor cells’ DNA oxidation, and repressing MMP-2 and MMP-9. CA and its derivatives have been reported to exhibit anti-carcinogenic properties against many cancer types. CA has indicated low intestinal absorption, low oral bioavailability in rats, and pitiable permeability across Caco-2 cells. In the present review, we have illustrated CA’s therapeutic potential, pharmacokinetics, and characteristics. The pharmacological effects of CA, the emphasis on in vitro and in vivo studies, and the existing challenges and prospects of CA for cancer treatment and prevention are discussed in this review.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kayenat Sheikh
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Anish Khan
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sabeeha Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Meraj Ansari
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, SAS Nagar Mohali, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Visweswara Rao Pasupuleti
- Department of Biomedical Sciences and Therapeutics, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Malaysia.,Department of Biochemistry, Faculty of Medicine and Health Sciences, Abdurrab University, Pekanbaru, Indonesia.,Centre for International Collaboration and Research, Reva University, Bangalore, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
11
|
Alam M, Ashraf GM, Sheikh K, Khan A, Ali S, Ansari MM, Adnan M, Pasupuleti VR, Hassan MI. Potential Therapeutic Implications of Caffeic Acid in Cancer Signaling: Past, Present, and Future. Front Pharmacol 2022. [DOI: 10.3389/fphar.2022.845871
expr 835330423 + 878857932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Caffeic acid (CA) has been present in many herbs, vegetables, and fruits. CA is a bioactive compound and exhibits various health advantages that are linked with its anti-oxidant functions and implicated in the therapy and prevention of disease progression of inflammatory diseases and cancer. The anti-tumor action of CA is attributed to its pro-oxidant and anti-oxidant properties. CA’s mechanism of action involves preventing reactive oxygen species formation, diminishing the angiogenesis of cancer cells, enhancing the tumor cells’ DNA oxidation, and repressing MMP-2 and MMP-9. CA and its derivatives have been reported to exhibit anti-carcinogenic properties against many cancer types. CA has indicated low intestinal absorption, low oral bioavailability in rats, and pitiable permeability across Caco-2 cells. In the present review, we have illustrated CA’s therapeutic potential, pharmacokinetics, and characteristics. The pharmacological effects of CA, the emphasis onin vitro and in vivostudies, and the existing challenges and prospects of CA for cancer treatment and prevention are discussed in this review.
Collapse
|
12
|
The Role of Epigenetic Modifications in Human Cancers and the Use of Natural Compounds as Epidrugs: Mechanistic Pathways and Pharmacodynamic Actions. Biomolecules 2022; 12:biom12030367. [PMID: 35327559 PMCID: PMC8945214 DOI: 10.3390/biom12030367] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer is a complex disease resulting from the genetic and epigenetic disruption of normal cells. The mechanistic understanding of the pathways involved in tumor transformation has implicated a priori predominance of epigenetic perturbations and a posteriori genetic instability. In this work, we aimed to explain the mechanistic involvement of epigenetic pathways in the cancer process, as well as the abilities of natural bioactive compounds isolated from medicinal plants (flavonoids, phenolic acids, stilbenes, and ketones) to specifically target the epigenome of tumor cells. The molecular events leading to transformation, angiogenesis, and dissemination are often complex, stochastic, and take turns. On the other hand, the decisive advances in genomics, epigenomics, transcriptomics, and proteomics have allowed, in recent years, for the mechanistic decryption of the molecular pathways of the cancerization process. This could explain the possibility of specifically targeting this or that mechanism leading to cancerization. With the plasticity and flexibility of epigenetic modifications, some studies have started the pharmacological screening of natural substances against different epigenetic pathways (DNA methylation, histone acetylation, histone methylation, and chromatin remodeling) to restore the cellular memory lost during tumor transformation. These substances can inhibit DNMTs, modify chromatin remodeling, and adjust histone modifications in favor of pre-established cell identity by the differentiation program. Epidrugs are molecules that target the epigenome program and can therefore restore cell memory in cancerous diseases. Natural products isolated from medicinal plants such as flavonoids and phenolic acids have shown their ability to exhibit several actions on epigenetic modifiers, such as the inhibition of DNMT, HMT, and HAT. The mechanisms of these substances are specific and pleiotropic and can sometimes be stochastic, and their use as anticancer epidrugs is currently a remarkable avenue in the fight against human cancers.
Collapse
|
13
|
Targeting Cancer Stem Cells by Dietary Agents: An Important Therapeutic Strategy against Human Malignancies. Int J Mol Sci 2021; 22:ijms222111669. [PMID: 34769099 PMCID: PMC8584029 DOI: 10.3390/ijms222111669] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/23/2021] [Accepted: 10/23/2021] [Indexed: 02/07/2023] Open
Abstract
As a multifactorial disease, treatment of cancer depends on understanding unique mechanisms involved in its progression. The cancer stem cells (CSCs) are responsible for tumor stemness and by enhancing colony formation, proliferation as well as metastasis, and these cells can also mediate resistance to therapy. Furthermore, the presence of CSCs leads to cancer recurrence and therefore their complete eradication can have immense therapeutic benefits. The present review focuses on targeting CSCs by natural products in cancer therapy. The growth and colony formation capacities of CSCs have been reported can be attenuated by the dietary agents. These compounds can induce apoptosis in CSCs and reduce tumor migration and invasion via EMT inhibition. A variety of molecular pathways including STAT3, Wnt/β-catenin, Sonic Hedgehog, Gli1 and NF-κB undergo down-regulation by dietary agents in suppressing CSC features. Upon exposure to natural agents, a significant decrease occurs in levels of CSC markers including CD44, CD133, ALDH1, Oct4 and Nanog to impair cancer stemness. Furthermore, CSC suppression by dietary agents can enhance sensitivity of tumors to chemotherapy and radiotherapy. In addition to in vitro studies, as well as experiments on the different preclinical models have shown capacity of natural products in suppressing cancer stemness. Furthermore, use of nanostructures for improving therapeutic impact of dietary agents is recommended to rapidly translate preclinical findings for clinical use.
Collapse
|
14
|
Wu HJ, Chu PY. Epigenetic Regulation of Breast Cancer Stem Cells Contributing to Carcinogenesis and Therapeutic Implications. Int J Mol Sci 2021; 22:ijms22158113. [PMID: 34360879 PMCID: PMC8348144 DOI: 10.3390/ijms22158113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Globally, breast cancer has remained the most commonly diagnosed cancer and the leading cause of cancer death among women. Breast cancer is a highly heterogeneous and phenotypically diverse group of diseases, which require different selection of treatments. Breast cancer stem cells (BCSCs), a small subset of cancer cells with stem cell-like properties, play essential roles in breast cancer progression, recurrence, metastasis, chemoresistance and treatments. Epigenetics is defined as inheritable changes in gene expression without alteration in DNA sequence. Epigenetic regulation includes DNA methylation and demethylation, as well as histone modifications. Aberrant epigenetic regulation results in carcinogenesis. In this review, the mechanism of epigenetic regulation involved in carcinogenesis, therapeutic resistance and metastasis of BCSCs will be discussed, and finally, the therapies targeting these biomarkers will be presented.
Collapse
Affiliation(s)
- Hsing-Ju Wu
- Department of Biology, National Changhua University of Education, Changhua 500, Taiwan;
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Medical Research, Chang Bing Show Chwan Memorial Hospital, Lukang Town, Changhua 505, Taiwan
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
- Correspondence: ; Tel.: +886-975611855; Fax: +886-47227116
| |
Collapse
|
15
|
Yang Y, Jin M, Dai Y, Shan W, Chen S, Cai R, Yang H, Tang L, Li L. Involvement and Targeted Intervention of Mortalin-Regulated Proteome Phosphorylated-Modification in Hepatocellular Carcinoma. Front Oncol 2021; 11:687871. [PMID: 34395254 PMCID: PMC8358780 DOI: 10.3389/fonc.2021.687871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/15/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES To reveal the mechanisms of the effects of mortalin in hepatocellular carcinoma (HCC) and to identify potential novel chemical inhibitors of mortalin. MATERIALS AND METHODS For the experiments, three HCC cell lines (HepG2 cells, Hep3B cells, and sorafenib-resistant HuH7 cells) and xenografted nude mice were used. For the clinical analysis, cohorts of 126 patients with HCC and 34 patients with advanced recurrent HCC receiving sorafenib therapy were examined. RESULTS Mortalin regulated the phosphorylation-modification of cancer-associated proteins and also regulated angiogenesis-related secretome to cause angiogenesis and sorafenib resistance in HCC cells. Two molecular mechanisms were identified. In one, via phosphatidylinositol 3-kinase (PI3K)/Akt signaling, mortalin regulated nuclear factor (NF)-κB and then activated vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor (VEGFR)2 and granulocyte-macrophage colony-stimulating factor (GM-CSF), leading to neovascularization. In the other, mortalin regulated PI3K/Akt/β-catenin and then regulated Bcl-XL and Bcl-2, leading to the antiapoptosis effect of HCC. Treatment of the sorafenib-resistant xenografts with sorafenib in combination with mortalin knockdown facilitated the sorafenib-mediated inhibition of tumor growth and angiogenesis and increased apoptosis. Mortalin was a potential risk factor for HCC, predicting poor prognosis and sorafenib resistance. Finally, we showed that caffeic acid (C9H8O4) could bind to and induce the ubiquitination-mediated degradation of mortalin, which in turn blocked the abovementioned signaling pathways, leading to the inhibition of angiogenesis and the reversal of sorafenib resistance. CONCLUSIONS Mortalin, which regulates the phosphorylation of cancer-associated proteins, caused angiogenesis and sorafenib resistance, and was a competitive risk factor for HCC. Caffeic acid can therefore be considered a novel chemical inhibitor that targets the action of mortalin and a potential treatment for HCC.
Collapse
Affiliation(s)
- Ye Yang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ming Jin
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yi Dai
- Department of General Surgery, The Affiliated Changzhou No. 2 Hospital of Nanjing Medical University, Changzhou, China
| | - Wenqi Shan
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Shuai Chen
- Department of General Surgery, The Affiliated Changzhou No. 2 Hospital of Nanjing Medical University, Changzhou, China
| | - Rong Cai
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Haojun Yang
- Department of General Surgery, The Affiliated Changzhou No. 2 Hospital of Nanjing Medical University, Changzhou, China
| | - Liming Tang
- Department of General Surgery, The Affiliated Changzhou No. 2 Hospital of Nanjing Medical University, Changzhou, China
| | - Lei Li
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Manogaran P, Umapathy D, Karthikeyan M, Venkatachalam K, Singaravelu A. Dietary Phytochemicals as a Potential Source for Targeting Cancer Stem Cells. Cancer Invest 2021; 39:349-368. [PMID: 33688788 DOI: 10.1080/07357907.2021.1894569] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The tumor microenvironment is composed of various types of cells that lead to tumor heterogeneity. In the middle of these populations, cancer stem cells play a vital role in the initiation and progression of cancer cells and are capable of self-renewal and differentiation processes. These cancer stem cells are resistant to conventional therapy such as chemotherapy and radiotherapy. To eradicate the cancer stem cells in the tumor environment, various natural product has been found in recent years. In this review, we have selected some of the natural products based on anticancer potential including targeting cancer cells and cancer stem cells. Further, this review explains the molecular mechanism of action of these natural products in various cancer stem cells. Therefore, targeting a multi-drug resistant cancer stem cell by natural products is a novel method to reduce drug resistance and adverse effect during conventional therapy.
Collapse
Affiliation(s)
- Prasath Manogaran
- Department of Biotechnology, Bharathiar University, Coimbatore, Tamilnadu, India
| | - Devan Umapathy
- Department of Biochemistry, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | | | - Karthikkumar Venkatachalam
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Anbu Singaravelu
- Department of PG and Research Department of Biochemistry, Sacred Heart College (Autonomous), Tirupattur, Tamilnadu, India
| |
Collapse
|
17
|
LncRNAs and microRNAs as Essential Regulators of Stemness in Breast Cancer Stem Cells. Biomolecules 2021; 11:biom11030380. [PMID: 33802575 PMCID: PMC7998729 DOI: 10.3390/biom11030380] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/13/2021] [Accepted: 02/22/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is an aggressive disease with a high incidence in women worldwide. Two decades ago, a controversial hypothesis was proposed that cancer arises from a subpopulation of “tumor initiating cells” or “cancer stem cells-like” (CSC). Today, CSC are defined as small subset of somatic cancer cells within a tumor with self-renewal properties driven by the aberrant expression of genes involved in the maintenance of a stemness-like phenotype. The understanding of the underlying cellular and molecular mechanisms involved in the maintenance of CSC subpopulation are fundamental in the development and persistence of breast cancer. Nowadays, the hypothesis suggests that genetic and epigenetic alterations give rise to breast cancer stem cells (bCSC), which are responsible for self-renewal, tumor growth, chemoresistance, poor prognosis and low survival in patients. However, the prominence of bCSC, as well as the molecular mechanisms that regulates and promotes the malignant phenotypes, are still poorly understood. The role of non-coding RNAs (ncRNAs), such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) acting as oncogenes or tumor suppressor genes has been recently highlighted by a plethora of studies in breast cancer. These ncRNAs positively or negatively impact on different signaling pathways that govern the cancer hallmarks associated with bCSC, making them attractive targets for therapy. In this review, we present a current summary of the studies on the pivotal roles of lncRNAs and microRNAs in the regulation of genes associated to stemness of bCSC.
Collapse
|
18
|
Hu J, Cao S, Zhang Z, Wang L, Wang D, Wu Q, Li L. Effects of caffeic acid on epigenetics in the brain of rats with chronic unpredictable mild stress. Mol Med Rep 2020; 22:5358-5368. [PMID: 33173990 PMCID: PMC7647007 DOI: 10.3892/mmr.2020.11609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
The present study hypothesized that caffeic acid (3,4-dihydroxycinnamic acid; CaA) may exert antidepressant-like effects in rats with chronic unpredictable mild stress via epigenetic mechanisms, such as DNA methylation and hydroxymethylation. The chronic unpredictable mild stress (CUMS) model was used to analyze the effects of CaA on behavioral phenotypes, and to evaluate the distribution of 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) in the hippocampus and prefrontal cortex using immunohistochemistry and immunofluorescence. mRNA levels of the genes encoding brain-derived neurotropic factor (BDNF) and catechol-O-methyltransferase (COMT), and key enzymes regulating DNA methylation [DNA methyltransferase (DNMT)1 and DNMT3A] and hydroxymethylation [Ten-eleven translocation (TET)1-3] were examined using quantitative (q)PCR. Furthermore, enrichment of 5mC and 5hmC at the promotor regions of the Bdnf and Comt genes was quantified using chromatin immunoprecipitation-qPCR. Behavioral data showed that CaA exerted a slight antidepressant-like effect. Bdnf and Comt genes showed differential expression patterns due to CUMS. CaA intervention induced different Dnmt1/Dnmt3a and Tet1/Tet2 mRNA levels in the hippocampus and prefrontal cortex, respectively. CaA regulated the ratio of 5mC/5hmC at the promotor region of the Bdnf and Comt genes and therefore influenced gene expression, which may be a valuable therapeutic option for major depressive disorder (MDD). In conclusion, there were epigenetic changes in the hippocampus and prefrontal cortex in CUMS rats, and CaA may function as a modulator of DNA methylation to regulate gene transcription, thus providing a mechanistic basis for the use of this phytochemical agent in the treatment of MDD.
Collapse
Affiliation(s)
- Jinye Hu
- Department of Health Inspection and Quarantine and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Shuyuan Cao
- Department of Health Inspection and Quarantine and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Zhan Zhang
- Department of Health Inspection and Quarantine and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Li Wang
- Department of Health Inspection and Quarantine and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Di Wang
- Department of Health Inspection and Quarantine and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Qian Wu
- Department of Health Inspection and Quarantine and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Lei Li
- Department of Health Inspection and Quarantine and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
19
|
Hernandes LC, Machado ART, Tuttis K, Ribeiro DL, Aissa AF, Dévoz PP, Antunes LMG. Caffeic acid and chlorogenic acid cytotoxicity, genotoxicity and impact on global DNA methylation in human leukemic cell lines. Genet Mol Biol 2020; 43:e20190347. [PMID: 32644097 PMCID: PMC7350414 DOI: 10.1590/1678-4685-gmb-2019-0347] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 05/29/2020] [Indexed: 12/20/2022] Open
Abstract
Dietary phenolic compounds such as caffeic and chlorogenic acid exert an antiproliferative effect and modulate the gene-specific DNA methylation status in human breast tumor cells, but it remains unclear whether they interfere with global DNA methylation in human leukemia cells. We examined whether caffeic and chlorogenic acid (1-250 µM) exert antitumor action in human promyelocytic leukemia cells (HL-60) and human acute T-cell leukemia cells (Jurkat). Caffeic and chlorogenic acid did not reduce cell viability in the two cell lines, as assessed using the neutral red uptake and MTT assays. These phenolic acids (1-100 μM) neither induced DNA damage (comet assay) nor increased the micronuclei frequency (micronucleus assay) in HL-60 and Jurkat cells, indicating that they were not genotoxic or mutagenic. Analysis of global DNA methylation levels using a 5-mC DNA ELISA kit revealed that chlorogenic acid at a non-cytotoxic concentration (100 μM) induced global DNA hypomethylation in Jurkat cells, but not in HL-60 cells, suggesting that it exerts a cell-specific effect. Caffeic acid did not change global DNA methylation. As other phenolic compounds, chlorogenic acid probably modulates DNA methylation by targeting DNA methyltransferases. The hypomethylating action of chlorogenic acid can be beneficial against hematological malignances whose pathogenic processes involve impairment of DNA methylation.
Collapse
Affiliation(s)
- Lívia Cristina Hernandes
- Universidade de São Paulo - USP, Faculdade de Ciências Farmacêuticas de Ribeirão Preto Ribeirão Preto, SP, Brazil
| | - Ana Rita Thomazela Machado
- Universidade de São Paulo - USP, Faculdade de Ciências Farmacêuticas de Ribeirão Preto Ribeirão Preto, SP, Brazil
| | - Katiuska Tuttis
- Universidade de São Paulo USP, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Diego Luís Ribeiro
- Universidade de São Paulo USP, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Alexandre Ferro Aissa
- Universidade de São Paulo - USP, Faculdade de Ciências Farmacêuticas de Ribeirão Preto Ribeirão Preto, SP, Brazil
| | - Paula Pícoli Dévoz
- Universidade de São Paulo - USP, Faculdade de Ciências Farmacêuticas de Ribeirão Preto Ribeirão Preto, SP, Brazil
| | - Lusânia Maria Greggi Antunes
- Universidade de São Paulo - USP, Faculdade de Ciências Farmacêuticas de Ribeirão Preto Ribeirão Preto, SP, Brazil
| |
Collapse
|
20
|
Wang M, Ye Q, Mao D, Li H. Research Progress in Liver-Regenerating Microenvironment and DNA Methylation in Hepatocellular Carcinoma: The Role of Traditional Chinese Medicine. Med Sci Monit 2020; 26:e920310. [PMID: 32144233 PMCID: PMC7077739 DOI: 10.12659/msm.920310] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The development, progression, recurrence, and metastasis of hepatocellular carcinoma (HCC) are closely associated with an abnormal liver-regenerating microenvironment (LRM). Therefore, preventing and reversing an abnormal LRM is a potential therapeutic strategy against HCC. Studies are increasingly focusing on the impact of regeneration, fibrosis, angiogenesis, inflammation, immunomodulation, and hepatic stem cells on HCC development and progression. As a key epigenetic mechanism, DNA methylation is extensively involved in regulating physiological and pathological pathways. In this review, we summarize recent findings on the role of DNA methylation in the fibrotic, angiogenic, inflammatory/immune, and stem cell microenvironments of HCC, and discuss new advances in Traditional Chinese Medicine (TCM) on influencing the abnormal LRM, so as to gain new insights into alleviating the abnormal LRM via regulating DNA methylation by TCM.
Collapse
Affiliation(s)
- Minggang Wang
- Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, China (mainland)
| | - Qianling Ye
- Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi, China (mainland)
| | - Dewen Mao
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi, China (mainland)
| | - Hanmin Li
- Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, China (mainland)
| |
Collapse
|
21
|
Therapeutic Potential of Plant Phenolic Acids in the Treatment of Cancer. Biomolecules 2020; 10:biom10020221. [PMID: 32028623 PMCID: PMC7072661 DOI: 10.3390/biom10020221] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/26/2020] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
Globally, cancer is the second leading cause of death. Different conventional approaches to treat cancer include chemotherapy or radiotherapy. However, these are usually associated with various deleterious effects and numerous disadvantages in clinical practice. In addition, there are increasing concerns about drug resistance. In the continuous search for safer and more effective treatments, plant-derived natural compounds are of major interest. Plant phenolics are secondary metabolites that have gained importance as potential anti-cancer compounds. Phenolics display a great prospective as cytotoxic anti-cancer agents promoting apoptosis, reducing proliferation, and targeting various aspects of cancer (angiogenesis, growth and differentiation, and metastasis). Phenolic acids are a subclass of plant phenolics, furtherly divided into benzoic and cinnamic acids, that are associated with potent anticancer abilities in various in vitro and in vivo studies. Moreover, the therapeutic activities of phenolic acids are reinforced by their role as epigenetic regulators as well as supporters of adverse events or resistance associated with conventional anticancer therapy. Encapsulation of phyto-substances into nanocarrier systems is a challenging aspect concerning the efficiency of natural substances used in cancer treatment. A summary of phenolic acids and their effectiveness as well as phenolic-associated advances in cancer treatment will be discussed in this review.
Collapse
|
22
|
Braicu C, Zimta AA, Gulei D, Olariu A, Berindan-Neagoe I. Comprehensive analysis of circular RNAs in pathological states: biogenesis, cellular regulation, and therapeutic relevance. Cell Mol Life Sci 2019; 76:1559-1577. [PMID: 30805658 PMCID: PMC11105679 DOI: 10.1007/s00018-019-03016-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 11/14/2018] [Accepted: 01/15/2019] [Indexed: 02/06/2023]
Abstract
Circular RNAs (circRNAs) are members of the non-coding transcriptome; however, some of them are translated into proteins. These transcripts have important roles in both physiological and pathological mechanisms due to their ability to directly influence cellular signaling pathways. Specifically, circRNAs are regulators of transcription, translation, protein interaction, and signal transduction. An increased knowledge within their area is observed over the last few years, concomitant with the development of next-generation sequencing techniques. circRNAs are mostly tissue and disease specific with the ability of specifically changing the biological behavior of cells. The altered expression profile is currently investigated as novel minimally invasive diagnosis/prognosis tool and also therapeutic target in human disease. The diagnosis approach is based on their level modification within pathological states, especially cancer, where circRNAs' therapies are intensively explored in anti-aging strategies, diabetes, cardiovascular diseases, and malignant pathologies, and are relying on the restoration of homeostatic profiles.
Collapse
Affiliation(s)
- Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania.
| | - Andreea-Alina Zimta
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Diana Gulei
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Andrei Olariu
- Nordlogic Software, 10-12, Rene Descartes Street, 400486, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania.
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania.
- Department of Functional Genomics and Experimental Pathology, "Prof. Dr. Ion Chiricuta" The Oncology Institute, 34-36 Republicii Street, 400015, Cluj-Napoca, Romania.
| |
Collapse
|
23
|
Biersack B. Relations between approved platinum drugs and non-coding RNAs in mesothelioma. Noncoding RNA Res 2018; 3:161-173. [PMID: 30809599 PMCID: PMC6260483 DOI: 10.1016/j.ncrna.2018.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 08/29/2018] [Indexed: 12/23/2022] Open
Abstract
Malignant mesothelioma diseases feature an increasing risk due to their severe forms and their association with asbestos exposure. Platinum(II) complexes such as cisplatin and carboplatin are clinically approved for the therapy of mesothelioma often in combination with antimetabolites such as pemetrexed or gemcitabine. It was observed that pathogenic properties of mesothelioma cells and the response of mesothelioma tumors towards platinum-based drugs are strongly influenced by non-coding RNAs, in particular, by small microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). These non-coding RNAs controlled drug sensitivity and the development of tumor resistance towards platinum drugs. An overview of the interactions between platinum drugs and non-coding RNAs is given and the influence of non-coding RNAs on platinum drug efficacy in mesothelioma is discussed. Suitable non-coding RNA-modulating agents with potentially beneficial effects on cisplatin treatment of mesothelioma diseases are mentioned. The understanding of mesothelioma diseases concerning the interactions of non-coding RNAs and platinum drugs will optimize existing therapy schemes and pave the way to new treatment options in future.
Collapse
Key Words
- ABC, ATP-binding cassette
- AKBA, 3-acetyl-11-keto-β-boswellic acid
- AKI, acute kidney injury
- Anticancer drugs
- Bcl-2, B-cell lymphoma 2
- CAF, cancer-associated fibroblast
- CBDCA, cyclobutane-1,1-dicarboxylate
- Carboplatin
- Cisplatin
- DADS, diallyl sulfide
- DHA, docosahexaenoic acid
- DIM, 3,3′-diindolylmethane
- DMPM, diffuse malignant peritoneal mesothelioma
- EGCG, epigallocatechin-3-gallate
- EMT, epithelial-mesenchymal transition
- HOTAIR, HOX transcript antisense RNA
- I3C, indole-3-carbinol
- Long non-coding RNA
- MALAT1, metastasis-associated lung adenocarcinoma transcript 1
- MPM, malignant pleural mesothelioma
- MRP1, multidrug resistance protein 1
- Mesothelioma
- MicroRNA
- NSCLC, non-small cell lung cancer
- NaB, sodium butyrate
- PDCD4, programmed cell death 4
- PEG, polyethylene glycole
- PEITC, phenethylisothiocyanate
- PTEN, phosphatase and tensin homolog
- RA, retinoic acid
- SAHA, suberoylanilide hydroxamic acid
- SFN, sulforaphane
- TNBC, triple-negative breast cancer
- TSA, trichostatin A
Collapse
|
24
|
Biersack B. Interplay of non-coding RNAs and approved antimetabolites such as gemcitabine and pemetrexed in mesothelioma. Noncoding RNA Res 2018; 3:213-225. [PMID: 30809600 PMCID: PMC6257890 DOI: 10.1016/j.ncrna.2018.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/03/2018] [Accepted: 11/03/2018] [Indexed: 12/13/2022] Open
Abstract
Gemcitabine and pemetrexed are clinically approved antimetabolites for the therapy of mesothelioma diseases. These drugs are often applied in combination with platinum complexes and other drugs. The activity of antimetabolites depended on the expression levels of certain non-coding RNAs, in particular, of small microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). The development of tumor resistance towards antimetabolites was regulated by non-coding RNAs. An overview of the interplay between gemcitabine/pemetrexed antimetabolites and non-coding RNAs in mesothelioma is provided. Further to this, various non-coding RNA-modulating agents are discussed which displayed positive effects on gemcitabine or pemetrexed treatment of mesothelioma diseases. A detailed knowledge of the connections of non-coding RNAs with antimetabolites will be constructive for the design of improved therapies in future.
Collapse
Key Words
- AKBA, 3-acetyl-11-keto-β-boswellic acid
- Anticancer drugs
- Bcl-2, B-cell lymphoma 2
- DADS, diallyl sulfide
- DHA, docosahexaenoic acid
- DIM, 3,3‘-diindolylmethane
- DMPM, diffuse malignant peritoneal mesothelioma
- EGCG, epigallocatechin-3-gallate
- EMT, epithelial-mesenchymal transition
- Gemcitabine
- HOTAIR, HOX transcript antisense RNA
- I3C, indole-3-carbinol
- Long non-coding RNA
- MALAT1, metastasis-associated lung adenocarcinoma transcript 1
- MPM, malignant pleural mesothelioma
- Mesothelioma
- MicroRNA
- NSCLC, non-small cell lung cancer
- NaB, sodium butyrate
- PDCD4, programmed cell death 4
- PEG, polyethylene glycole
- PEITC, phenethylisothiocyanate
- PTEN, phosphatase and tensin homolog
- Pemetrexed
- RA, retinoic acid
- SAHA, suberoylanilide hydroxamic acid
- SFN, sulforaphane
- TSA, trichostatin A
Collapse
|
25
|
Cruz-Lozano M, González-González A, Marchal JA, Muñoz-Muela E, Molina MP, Cara FE, Brown AM, García-Rivas G, Hernández-Brenes C, Lorente JA, Sanchez-Rovira P, Chang JC, Granados-Principal S. Hydroxytyrosol inhibits cancer stem cells and the metastatic capacity of triple-negative breast cancer cell lines by the simultaneous targeting of epithelial-to-mesenchymal transition, Wnt/β-catenin and TGFβ signaling pathways. Eur J Nutr 2018; 58:3207-3219. [DOI: 10.1007/s00394-018-1864-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/14/2018] [Indexed: 02/07/2023]
|
26
|
Lu M, Dai Y, Xu M, Zhang C, Ma Y, Gao P, Teng M, Jiao K, Huang G, Zhang J, Yang Y, Chu Z. The Attenuation of 14-3-3ζ is Involved in the Caffeic Acid-Blocked Lipopolysaccharide-Stimulated Inflammatory Response in RAW264.7 Macrophages. Inflammation 2018; 40:1753-1760. [PMID: 28688098 DOI: 10.1007/s10753-017-0618-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Inflammation plays important roles in the initiation and progress of many diseases. Caffeic acid (CaA) is a naturally occurring hydroxycinnamic acid derivative, which shows hypotoxicity and diverse biological functions, including anti-inflammation. The molecular mechanisms involved in the CaA-inhibited inflammatory response are very complex; generally, the down-regulated phosphorylation of such important transcriptional factors, for example, nuclear factor κB (NF-κB) and signal transducers and activators of transcription-3 (STAT-3), plays an important role. Here, we found that in RAW264.7 macrophage cells, CaA blocked lipopolysaccharide (LPS)-stimulated inflammatory response by attenuating the expression of 14-3-3ζ (a phosphorylated protein regulator). Briefly, the increased expression of 14-3-3ζ was involved in the LPS-induced inflammatory response. CaA blocked the LPS-elevated 14-3-3ζ via attenuating the LPS-induced tumor necrosis factor-α (TNF-α) secretion and via enhancing the 14-3-3ζ ubiquitination. These processes inhibited the LPS-induced activation (phosphorylation) of NF-κB and STAT-3, in turn blocked the transcriptional activation of inducible NO synthase (iNOS), interleukin-6 (IL-6), and TNF-α, and finally attenuated the productions of nitric oxide (NO), IL-6, and TNF-α. By understanding a novel mechanism whereby CaA inhibited the 14-3-3ζ, our study expanded the understanding of the molecular mechanisms involved in the anti-inflammation potential induced by CaA.
Collapse
Affiliation(s)
- Ming Lu
- The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Yi Dai
- The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Miao Xu
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chi Zhang
- The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Yuhong Ma
- The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Ping Gao
- The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Mengying Teng
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Kailin Jiao
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Guangming Huang
- The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Jianping Zhang
- The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China.
| | - Ye Yang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Zhiping Chu
- The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China.
| |
Collapse
|
27
|
Yu Y, Luo W, Yang ZJ, Chi JR, Li YR, Ding Y, Ge J, Wang X, Cao XC. miR-190 suppresses breast cancer metastasis by regulation of TGF-β-induced epithelial-mesenchymal transition. Mol Cancer 2018; 17:70. [PMID: 29510731 PMCID: PMC5838994 DOI: 10.1186/s12943-018-0818-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/26/2018] [Indexed: 01/23/2023] Open
Abstract
Background Breast cancer is the most common cancer among women worldwide and metastasis is the leading cause of death among patients with breast cancer. The transforming growth factor-β (TGF-β) pathway plays critical roles during breast cancer epithelial–mesenchymal transition (EMT) and metastasis. SMAD2, a positive regulator of TGF-β signaling, promotes breast cancer metastasis through induction of EMT. Methods The expression of miR-190 and SMAD2 in breast cancer tissues, adjacent normal breast tissues and cell lines were determined by RT-qPCR. The protein expression levels and localization were analyzed by western blotting and immunofluorescence. ChIP and dual-luciferase report assays were used to validate the regulation of ZEB1-miR-190-SMAD2 axis. The effect of miR-190 on breast cancer progression was investigated both in vitro and in vivo. Results miR-190 down-regulation is required for TGF-β-induced EMT. miR-190 suppresses breast cancer metastasis both in vitro and in vivo by targeting SMAD2. miR-190 expression is down-regulated and inversely correlates with SMAD2 in breast cancer samples, and its expression level was associated with outcome in patients with breast cancer. Furthermore, miR-190 is transcriptionally regulated by ZEB1. Conclusions Our data uncover the ZEB1-miR-190-SMAD2 axis and provide a mechanism to explain the TGF-β network in breast cancer metastasis. Electronic supplementary material The online version of this article (10.1186/s12943-018-0818-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yue Yu
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Wei Luo
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Zheng-Jun Yang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Jiang-Rui Chi
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Yun-Rui Li
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Yu Ding
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Jie Ge
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Xin Wang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Xu-Chen Cao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China. .,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
28
|
Romagnolo DF, Daniels KD, Grunwald JT, Ramos SA, Propper CR, Selmin OI. Epigenetics of breast cancer: Modifying role of environmental and bioactive food compounds. Mol Nutr Food Res 2017; 60:1310-29. [PMID: 27144894 DOI: 10.1002/mnfr.201501063] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 04/24/2016] [Accepted: 04/26/2016] [Indexed: 12/12/2022]
Abstract
SCOPE Reduced expression of tumor suppressor genes (TSG) increases the susceptibility to breast cancer. However, only a small percentage of breast tumors is related to family history and mutational inactivation of TSG. Epigenetics refers to non-mutational events that alter gene expression. Endocrine disruptors found in foods and drinking water may disrupt epigenetically hormonal regulation and increase breast cancer risk. This review centers on the working hypothesis that agonists of the aromatic hydrocarbon receptor (AHR), bisphenol A (BPA), and arsenic compounds, induce in TSG epigenetic signatures that mirror those often seen in sporadic breast tumors. Conversely, it is hypothesized that bioactive food components that target epigenetic mechanisms protect against sporadic breast cancer induced by these disruptors. METHODS AND RESULTS This review highlights (i) overlaps between epigenetic signatures placed in TSG by AHR-ligands, BPA, and arsenic with epigenetic alterations associated with sporadic breast tumorigenesis; and (ii) potential opportunities for the prevention of sporadic breast cancer with food components that target the epigenetic machinery. CONCLUSIONS Characterizing the overlap between epigenetic signatures elicited in TSG by endocrine disruptors with those observed in sporadic breast tumors may afford new strategies for breast cancer prevention with specific bioactive food components or diet.
Collapse
Affiliation(s)
- Donato F Romagnolo
- Department of Nutritional Sciences, The University of Arizona, Tucson, AZ, USA.,The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Kevin D Daniels
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Jonathan T Grunwald
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Stephan A Ramos
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Catherine R Propper
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Ornella I Selmin
- Department of Nutritional Sciences, The University of Arizona, Tucson, AZ, USA.,The University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
29
|
Shao F, Sun H, Deng CX. Potential therapeutic targets of triple-negative breast cancer based on its intrinsic subtype. Oncotarget 2017; 8:73329-73344. [PMID: 29069872 PMCID: PMC5641215 DOI: 10.18632/oncotarget.20274] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/06/2017] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subgroup of human breast cancer, which is characterized as estrogen receptor (ER) negative, progesterone receptor (PR) negative, and human epidermal growth factor receptor 2 (HER2) negative. TNBC is the most difficult breast cancer subgroup to treat, due to its unresponsiveness to current clinical targeted therapies, high rate of recurrence, and poor prognosis. Thus, there is an urgent medical need to identify therapeutic targets and develop more effective stratified medicine for the treatment of TNBC. Here we review the potential therapeutic targets for TNBC based on its intrinsic subtype. We also review the aberrant activated signals found in different subgroups of TNBC, including androgen receptor (AR) and PI3K/AKT/mTOR, Notch, Wnt/β-catenin, Hedge-hog, and TGF-β signaling pathways, which play essential roles in multiple development stages of TNBC. The careful analysis of these signaling pathways and therapeutic targets would have significant impact on the drug development and clinical trials, leading to effective therapies for this deadly disease.
Collapse
Affiliation(s)
- Fangyuan Shao
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Heng Sun
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
30
|
Zhang Z, Wang D, Qiao S, Wu X, Cao S, Wang L, Su X, Li L. Metabolic and microbial signatures in rat hepatocellular carcinoma treated with caffeic acid and chlorogenic acid. Sci Rep 2017; 7:4508. [PMID: 28674386 PMCID: PMC5495756 DOI: 10.1038/s41598-017-04888-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/22/2017] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) treatment remains lack of effective chemopreventive agents, therefore it is very attractive and urgent to discover novel anti-HCC drugs. In the present study, the effects of chlorogenic acid (ChA) and caffeic acid (CaA) on HCC induced by diethylnitrosamine (DEN) were evaluated. ChA or CaA could reduce the histopathological changes and liver injury markers, such as alanine transarninase, aspartate aminotransferase, alkaline phosphatase, total bile acid, total cholesterol, high density lipoprotein cholesterol and low density lipoprotein cholesterol. The underlying mechanisms were investigated by a data integration strategy based on correlation analyses of metabonomics data and 16 S rRNA gene sequencing data. ChA or CaA could inhibit the increase of Rumincoccaceae UCG-004 and reduction of Lachnospiraceae incertae sedis, and Prevotella 9 in HCC rats. The principal component analysis and partial least squares discriminant analysis were applied to reveal the metabolic differences among these groups. 28 different metabolites showed a trend to return to normal in both CaA and ChA treatment. Among them, Bilirubin, L-Tyrosine, L-Methionine and Ethanolamine were correlated increased Rumincoccaceae UCG-004 and decreased of Lachnospiraceae incertae sedis and Prevotella 9. These correlations could be identified as metabolic and microbial signatures of HCC onset and potential therapeutic targets.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, P. R. China
| | - Di Wang
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, P. R. China
| | - Shanlei Qiao
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, P. R. China
| | - Xinyue Wu
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, P. R. China
| | - Shuyuan Cao
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, P. R. China
| | - Li Wang
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, P. R. China
| | - Xiaojian Su
- Nanjing entry-exit inspection and quarantine bureau, 110 Jiangjun Avenue, Nanjing, 211106, Jiangsu, P.R. China
| | - Lei Li
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, P. R. China.
| |
Collapse
|
31
|
Braicu C, Mehterov N, Vladimirov B, Sarafian V, Nabavi SM, Atanasov AG, Berindan-Neagoe I. Nutrigenomics in cancer: Revisiting the effects of natural compounds. Semin Cancer Biol 2017; 46:84-106. [PMID: 28676460 DOI: 10.1016/j.semcancer.2017.06.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 06/04/2017] [Accepted: 06/23/2017] [Indexed: 01/06/2023]
Abstract
Nutrigenomics effects have an important role in the manipulation of dietary components for human benefit, particularly in cancer prevention or treatment. The impact of dietary components, including phytochemicals, is largely studied by nutrigenomics, looking at the gene expression and molecular mechanisms interacting with bioactive compounds and nutrients, based on new 'omics' technologies. The high number of preclinical studies proves the relevant role of nutrigenomics in cancer management. By deciphering the network of nutrient-gene connections associated with cancer, relevant data will be transposed as therapeutic interventions for this devastating pathology and for fulfilling the concept of personalized nutrition. All these are presented under the nutrigenomics canopy for a better comprehension of the relation between ingested phytochemicals and chemoprevention or chemotherapy. The profits from the nutrigenomics progress, with a particular focus on the coding and noncoding genes related to the exposure of natural compounds need to be validated. A precise attention receives the evaluation of the role of natural compounds in tandem with conventional therapy using genomic approaches, with emphasis on the capacity to inhibit drug resistance mechanisms. All these relevant nutrigenomics aspects are summarized in the present review paper. It is concluded that further nutrigenomics studies are required to improve our understanding related to the complex mechanisms of action of the natural compounds and for their appropriate application as gears in cancer therapy.
Collapse
Affiliation(s)
- Cornelia Braicu
- Research Center for Functional Genomics and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 40015 Cluj-Napoca, Romania
| | - Nikolay Mehterov
- Department of Medical Biology, Faculty of Medicine, Medical University-Plovdiv, 15-А Vassil Aprilov Blvd., Plovdiv 4000, Bulgaria; Technological Center for Emergency Medicine, 15-А Vassil Aprilov Blvd., Plovdiv 4000, Bulgaria; Center of Plant Systems Biology and Biotechnology, 139, Ruski Blvd., Plovdiv 4000, Bulgaria
| | - Boyan Vladimirov
- Department of Maxillofacial Surgery, Faculty of Dental Medicine, Medical University-Plovdiv, 3 Hristo Botev Blvd., Plovdiv 4000, Bulgaria; Clinic of Maxillofacial Surgery, University Hospital St. George, 66 Peshtersko Shosse Blvd., Plovdiv 4002, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Faculty of Medicine, Medical University-Plovdiv, 15-А Vassil Aprilov Blvd., Plovdiv 4000, Bulgaria; Technological Center for Emergency Medicine, 15-А Vassil Aprilov Blvd., Plovdiv 4000, Bulgaria
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Sheikh Bahaei St., P.O. Box 19395, 5487 Tehran, Iran
| | - Atanas G Atanasov
- Department of Pharmacognosy, University of Vienna, Althanstrasse 14, Vienna 1090, Austria; Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Postępu 36A Street, 05-552, Jastrzebiec, Poland; Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 40015 Cluj-Napoca, Romania; MEDFUTURE -Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, 23 Marinescu Street, 40015, Romania; Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Republici 34 Street, 400015 Cluj-Napoca, Romania.
| |
Collapse
|
32
|
Budisan L, Gulei D, Zanoaga OM, Irimie AI, Sergiu C, Braicu C, Gherman CD, Berindan-Neagoe I. Dietary Intervention by Phytochemicals and Their Role in Modulating Coding and Non-Coding Genes in Cancer. Int J Mol Sci 2017; 18:ijms18061178. [PMID: 28587155 PMCID: PMC5486001 DOI: 10.3390/ijms18061178] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/20/2017] [Accepted: 05/24/2017] [Indexed: 12/13/2022] Open
Abstract
Phytochemicals are natural compounds synthesized as secondary metabolites in plants, representing an important source of molecules with a wide range of therapeutic applications. These natural agents are important regulators of key pathological processes/conditions, including cancer, as they are able to modulate the expression of coding and non-coding transcripts with an oncogenic or tumour suppressor role. These natural agents are currently exploited for the development of therapeutic strategies alone or in tandem with conventional treatments for cancer. The aim of this paper is to review the recent studies regarding the role of these natural phytochemicals in different processes related to cancer inhibition, including apoptosis activation, angiogenesis and metastasis suppression. From the large palette of phytochemicals we selected epigallocatechin gallate (EGCG), caffeic acid phenethyl ester (CAPE), genistein, morin and kaempferol, due to their increased activity in modulating multiple coding and non-coding genes, targeting the main hallmarks of cancer.
Collapse
Affiliation(s)
- Liviuta Budisan
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy "Iuliu-Hatieganu", 400012 Cluj-Napoca, Romania.
| | - Diana Gulei
- MEDFUTURE-Research Center for Advanced Medicine, University of Medicine and Pharmacy "Iuliu-Hatieganu", 400012 Cluj-Napoca, Romania.
| | - Oana Mihaela Zanoaga
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy "Iuliu-Hatieganu", 400012 Cluj-Napoca, Romania.
| | - Alexandra Iulia Irimie
- Department of Prosthodontics and Dental Materials, Faculty of Dental Medicine, University of Medicine and Pharmacy "Iuliu Hatieganu", 23 Marinescu Street, 400012 Cluj-Napoca, Romania.
| | - Chira Sergiu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy "Iuliu-Hatieganu", 400012 Cluj-Napoca, Romania.
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy "Iuliu-Hatieganu", 400012 Cluj-Napoca, Romania.
| | - Claudia Diana Gherman
- Surgical Clinic II, 4-6 Clinicilor Street, 400006 Cluj-Napoca, Romania.
- Department of Surgery, University of Medicine and Pharmacy "Iuliu Haţieganu", 8 Victor Babes Street, 400012 Cluj-Napoca, Romania.
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy "Iuliu-Hatieganu", 400012 Cluj-Napoca, Romania.
- MEDFUTURE-Research Center for Advanced Medicine, University of Medicine and Pharmacy "Iuliu-Hatieganu", 400012 Cluj-Napoca, Romania.
- Department of Functional Genomics and Experimental Pathology, Oncological Institute "Prof. Dr. Ion Chiricuţă", 400015 Cluj-Napoca, Romania.
| |
Collapse
|
33
|
Lee H, Vilian ATE, Kim JY, Chun MH, Suh JS, Seo HH, Cho SH, Shin IS, Kim SJ, Park SH, Han YK, Lee JH, Huh YS. Design and development of caffeic acid conjugated with Bombyx mori derived peptide biomaterials for anti-aging skin care applications. RSC Adv 2017. [DOI: 10.1039/c7ra04138a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This study examined the significant features of caffeic acid conjugated with peptide (APPPKK) to determine if it meets the requirements for a cosmetic anti-aging biomolecule.
Collapse
|
34
|
Yuan X, Kong J, Ma Z, Li N, Jia R, Liu Y, Zhou F, Zhan Q, Liu G, Gao S. KDM4C, a H3K9me3 Histone Demethylase, is Involved in the Maintenance of Human ESCC-Initiating Cells by Epigenetically Enhancing SOX2 Expression. Neoplasia 2016; 18:594-609. [PMID: 27742014 PMCID: PMC5035342 DOI: 10.1016/j.neo.2016.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/16/2016] [Accepted: 08/24/2016] [Indexed: 01/16/2023] Open
Abstract
Our studies investigating the existence of tumor-initiating cell (TIC) populations in human esophageal squamous cell carcinoma (ESCC) had identified a subpopulation of cells isolated from ESCC patient-derived tumor specimens marked by an ALDHbri+ phenotype bear stem cell-like features. Importantly, KDM4C, a histone demethylase was enhanced in ALDHbri+ subpopulation, suggesting that strategies interfering with KDM4C may be able to target these putative TICs. In the present study, by genetic and chemical means, we demonstrated that, KDM4C blockade selectively decreased the ESCC ALDHbri+ TICs population in vitro and specifically targeted the TICs in ALDHbri+-derived xenograft, retarding engraftment. Subsequent studies of the KDM4C functional network identified a subset of pluripotency-associated genes (PAGs) and aldehyde dehydrogenase family members to be preferentially down-regulated in KDM4C inhibited ALDHbri+ TICs. We further supported a model in which KDM4C maintains permissive histone modifications with a low level of H3K9 methylation at the promoters of several PAGs. Moreover, ectopic expression of SOX2 restored KDM4C inhibition-dependent ALDHbri+ TIC properties. We further confirmed these findings by showing that the cytoplasmic and nuclear KDM4C staining increased with adverse pathologic phenotypes and poor patient survival. Such staining pattern of intracellular KDM4C appeared to overlap with the expression of SOX2 and ALDH1. Collectively, our findings provided the insights into the development of novel therapeutic strategies based on the inhibition of KDM4C pathway for the eliminating of ESCC TIC compartment.
Collapse
Affiliation(s)
- Xiang Yuan
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| | - Jinyu Kong
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| | - Zhikun Ma
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| | - Na Li
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| | - Ruinuo Jia
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| | - Yiwen Liu
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| | - Fuyou Zhou
- Department of Thoracic Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking, China.
| | - Gang Liu
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China; Barbara Ann Karmanos Cancer Institute and Department of Oncology, Wayne State University, 4100 John R, Detroit, MI 48201.
| | - Shegan Gao
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
35
|
Chen L, Li Y, Yin W, Shan W, Dai J, Yang Y, Li L. Combination of chlorogenic acid and salvianolic acid B protects against polychlorinated biphenyls-induced oxidative stress through Nrf2. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 46:255-263. [PMID: 27513569 DOI: 10.1016/j.etap.2016.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/30/2016] [Accepted: 08/03/2016] [Indexed: 06/06/2023]
Abstract
Caffeic acid derivatives (CADs) are well-known phytochemicals with multiple physiological and pharmacological activities. This study aimed to investigate the combined protective effects of CADs on PCB126-induced liver damages and oxidative stress in mice. Here, we used chemiluminescence and chose chlorogenic acid (CGA), salvianolic acid B (Sal B) as the best antioxidants. Then, mice were intragastrically administered with 60mg/kg/d CGA, Sal B, and CGA plus Sal B (1:1) for 3 weeks before exposing to 0.05mg/kg/d PCB126 for 2 weeks. We found that pretreatment with CGA, Sal B, and CGA plus Sal B effectively attenuated liver injury and cytotoxicity caused by PCB126, but improved the expressions of superoxide dismutase (SOD), glutathione reduced (GSH), heme oxygenase-1 (HO-1) and nuclear factor E2-related factor 2 (Nrf2), CGA plus Sal B especially, was found to have the best effects that indicated a synergetic protective effect. Taken together, as the Nrf2 regulates the cyto-protective response by up-regulating the expression of antioxidant genes, we suggested that CGA plus Sal B had a combined protection on PCB126-induced tissue damages and that the Nrf2 signaling might be involved.
Collapse
Affiliation(s)
- Lijun Chen
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Yuan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Wenqin Yin
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Wenqi Shan
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Jinfeng Dai
- The Key Laboratory of Modern Toxicology, Ministry of Education, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Ye Yang
- The Key Laboratory of Modern Toxicology, Ministry of Education, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Lei Li
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing 211166, People's Republic of China.
| |
Collapse
|
36
|
Biersack B. Current state of phenolic and terpenoidal dietary factors and natural products as non-coding RNA/microRNA modulators for improved cancer therapy and prevention. Noncoding RNA Res 2016; 1:12-34. [PMID: 30159408 PMCID: PMC6096431 DOI: 10.1016/j.ncrna.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/20/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023] Open
Abstract
The epigenetic regulation of cancer cells by small non-coding RNA molecules, the microRNAs (miRNAs), has raised particular interest in the field of oncology. These miRNAs play crucial roles concerning pathogenic properties of cancer cells and the sensitivity of cancer cells towards anticancer drugs. Certain miRNAs are responsible for an enhanced activity of drugs, while others lead to the formation of tumor resistance. In addition, miRNAs regulate survival and proliferation of cancer cells, in particular of cancer stem-like cells (CSCs), that are especially drug-resistant and, thus, cause tumor relapse in many cases. Various small molecule compounds were discovered that target miRNAs that are known to modulate tumor aggressiveness and drug resistance. This review comprises the effects of naturally occurring small molecules (phenolic compounds and terpenoids) on miRNAs involved in cancer diseases.
Collapse
Key Words
- 1,25-D, 1,25-dihydroxyvitamin D3
- 18-AGA, 18α-glycyrrhetinic acid
- 3,6-DHF, 3,6-dihydroxyflavone
- AKBA, 3-acetyl-11-keto-β-boswellic acid
- Anticancer drugs
- CAPE, caffeic acid phenethyl ester
- CDODA-Me, methyl 2-cyano-3,11-dioxo-18β-olean-1,12-dien-30-oate
- Dox, doxorubicin
- EGCG, (−)-epigallocatechin-3-O-gallate
- MicroRNA
- PEG, polyethylene glycol
- PPAP, polycyclic polyprenylated acylphloroglucinol
- Polyphenols
- RA, retinoic acid
- ROS, reactive oxygen species
- TQ, thymoquinone
- Terpenes
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
| |
Collapse
|