1
|
Lin W, Zhao Z, Du W, Ni Z, Pan C, Fang P, Li J, ZhuGe L, Jin S. Interferon-Gamma-Inducible Protein 16 Inhibits Hepatocellular Carcinoma via Interferon Regulatory Factor 3 on Chemosensitivity. Dig Dis Sci 2024; 69:491-501. [PMID: 38170337 DOI: 10.1007/s10620-023-08175-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/29/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND AND AIM Previous reports have suggested IFI16 as a tumor suppressor in hepatocellular carcinoma (HC). Nonetheless, the biological significance of IFI16 and its mechanism concerning resistance to cisplatin (DDP) in HC requires further exploration. METHODS Samples of tumor and corresponding para-carcinoma tissues were acquired from patients with HC. Furthermore, DDP-resistant cell lines of HC, specifically HCC, Huh7 and Hepatoblastoma, HepG3, were generated by gradually increasing the concentration of DDP. Cell apoptosis and DNA damage were evaluated by utilizing flow cytometry assay and TUNEL staining. The interaction between IFI16 and interferon regulatory factor 3 (IRF3) proteins were analyzed using Co-Immunoprecipitation (Co-IP) assay. In vivo assays were conducted by establishing HC subcutaneous xenograft tumor models. RESULTS The study found a reduction in IFI16 expression in both HC tissues and DDP-resistant HC cell lines. The binding of IFI16 to IRF3 regulated DNA damage-associated markers in vitro. Overexpression of IFI16 heightened the susceptibility of DDP-induced apoptosis and DNA damage, which was counteracted by IRF3 knockdown, while strengthened by IRF3 overexpression. Moreover, overexpression of IFI16 diminished in vivo DDP-resistant HC tumorigenicity. CONCLUSION In summary, our findings suggest that IFI16 serves as a tumor suppressor in HC by promoting DNA damage via its interaction with IRF3, thereby reversing DDP resistance.
Collapse
Affiliation(s)
- Wei Lin
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China.
| | - Zhiguang Zhao
- Department of Pathology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenjun Du
- Department of Liver Diseases, Shandong Public Health Clinical Center, Shangdong University, Jinan, Shangdong, China
| | - Zhonglin Ni
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenwei Pan
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China
| | - Peipei Fang
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China
| | - Jie Li
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China
| | - Lu ZhuGe
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China
| | - Shuanghong Jin
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China
| |
Collapse
|
2
|
Li ZQ, Zhang GS, Liu RQ, Shuai SY, Hu PY, Zheng Q, Xiao SH. Anti-Glioma Effects of Ligustilide or n-Butylphthalide on Their Own and the Synergistic Effects with Temozolomide via PI3K/Akt Signaling Pathway. Onco Targets Ther 2023; 16:983-994. [PMID: 38021448 PMCID: PMC10676728 DOI: 10.2147/ott.s432901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Background Ligustilide (LIG) and n-butylphthalide (NBP) have neuroprotective effects in cerebral ischemia; however, their roles in gliomas are not well-known.This study aimed to explore the anti-glioma effects of LIG and NBP individually and the synergistic effects of temozolomide (TMZ) via the PI3K/Akt Signaling Pathway. Materials and Methods Cytotoxicity of LIG and NBP alone and in combination with TMZ in U251 cells was determined using the CCk-8. The effect of compounds alone or in combination on cell migration was detected using the wound healing assay, and the invasion was evaluated by transwell assays, respectively. Cell apoptosis was quantified by flow cytometry and the changed expressions of proteins were detected by Western blotting. Results The results showed that LIG and NBP significantly inhibited the growth of U251 cells at concentrations of 4-10 µg/mL and 1.5-6 µg/mL in a dose-dependent manner (p<0.05, p<0.01). The combination of 20 µg/mL TMZ with LIG in the concentration range of 4-10 µg/mL or with NBP of 0.5-6 µg/mlachieved synergistic effect towardsU251 cells. LIG and NBP, alone or in combination with TMZ, markedly inhibited cell invasion (p< 0.001) and enhanced apoptosis (p< 0.05). The combination of TMZ with LIG or NBP markedly inhibited cell migration (p< 0.001). Western blot analysis showed that LIG, NBP, and TMZ, alone and in combination, significantly decreased the expression of Bcl-2, p-PI3K, and p-Akt, and increased the expression of Bax. Conclusion Both LIG and NBP exert anti-glioma effects on their own through the PI3K/Akt pathway and enhance TMZ-mediated anti-glioma efficiency via the same pathway.
Collapse
Affiliation(s)
- Zi-Qi Li
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Guo-Song Zhang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Ri-Qun Liu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Shu-Yuan Shuai
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Peng-Yi Hu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Shu-Hua Xiao
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| |
Collapse
|
3
|
Al-Kabariti AY, Abbas MA. Progress in the Understanding of Estrogen Receptor Alpha Signaling in Triple-Negative Breast Cancer: Reactivation of Silenced ER-α and Signaling through ER-α36. Mol Cancer Res 2023; 21:1123-1138. [PMID: 37462782 DOI: 10.1158/1541-7786.mcr-23-0321] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/21/2023] [Accepted: 07/14/2023] [Indexed: 11/02/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive tumor that accounts for approximately 15% of total breast cancer cases. It is characterized by poor prognosis and high rate of recurrence compared to other types of breast cancer. TNBC has a limited range of treatment options that include chemotherapy, surgery, and radiation due to the absence of estrogen receptor alpha (ER-α) rendering hormonal therapy ineffective. However, possible targets for improving the clinical outcomes in TNBC exist, such as targeting estrogen signaling through membranous ER-α36 and reactivating silenced ER-α. It has been shown that epigenetic drugs such as DNA methyltransferase and histone deacetylase inhibitors can restore the expression of ER-α. This reactivation of ER-α, presents a potential strategy to re-sensitize TNBC to hormonal therapy. Also, this review provides up-to-date information related to the direct involvement of miRNA in regulating the translation of ER-α mRNA. Specific epi-miRNAs can regulate ER-α expression indirectly by post-transcriptional targeting of mRNAs of enzymes that are involved in DNA methylation and histone deacetylation. Furthermore, ER-α36, an alternative splice variant of ER-α66, is highly expressed in ER-negative breast tumors and activates MAPK/ERK pathway, promoting cell proliferation, escaping apoptosis, and enhancing metastasis. In the future, these recent advances may be helpful for researchers working in the field to obtain novel treatment options for TNBC, utilizing epigenetic drugs and epi-miRNAs that regulate ER-α expression. Also, there is some evidence to suggest that drugs that decrease the expression of ER-α36 may be effective in treating TNBC.
Collapse
Affiliation(s)
- Aya Y Al-Kabariti
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman, Jordan
| | - Manal A Abbas
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman, Jordan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| |
Collapse
|
4
|
Wu YY, Xu YM, Lau ATY. Epigenetic effects of herbal medicine. Clin Epigenetics 2023; 15:85. [PMID: 37179342 PMCID: PMC10183144 DOI: 10.1186/s13148-023-01481-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 04/08/2023] [Indexed: 05/15/2023] Open
Abstract
Epigenetic memory is essential for life that governs the predefined functional features of cells. Recent evidence has indicated that the epigenetic modification provides a potential link to gene expression changes that may be involved in the development of various chronic diseases, and targeting the epigenome becomes a plausible method for treating diseases. Traditional herbal medicine has gradually entered the vision of researchers due to its low toxicity and its effectiveness in treating diseases. As a matter of fact, researchers found that the possessed epigenetic modification capacity of herbal medicine had the ability to combat the progression of the disease, such as various types of cancer, diabetes, inflammation, amnesia, liver fibrosis, asthma, and hypertension-induced renal injury. Studies on the epigenetic effects of herbal medicine will provide valuable insights into the molecular mechanisms of human diseases, which may lead to new therapeutic approaches and diagnoses. Thus, this review summarized the impact of herbal medicine and its bioactive components on disease epigenome as examples of how utilization of epigenetic plasticity could be useful as the basis for the future development of targeted therapies in chronic diseases.
Collapse
Affiliation(s)
- Yu-Yao Wu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China.
| |
Collapse
|
5
|
Ma TL, Chang KF, Huang XF, Lai HC, Hsiao CY, Tsai NM. Angelica sinensis extract induces telomere dysfunction, cell cycle arrest, and mitochondria-mediated apoptosis in human glioblastoma cells. CHINESE J PHYSIOL 2023; 66:119-128. [PMID: 37322622 DOI: 10.4103/cjop.cjop-d-23-00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023] Open
Abstract
Glioblastoma (GB) is one of the most aggressive and malignant tumors of the central nervous system. Conventional treatment for GB requires surgical resection followed by radiotherapy combined with temozolomide chemotherapy; however, the median survival time is only 12-15 months. Angelica sinensis Radix (AS) is commonly used as a traditional medicinal herb or a food/dietary supplement in Asia, Europe, and North America. This study aimed to investigate the effect of AS-acetone extract (AS-A) on the progression of GB and the potential mechanisms underlying its effects. The results indicated that AS-A used in this study showed potency in growth inhibition of GB cells and reduction of telomerase activity. In addition, AS-A blocked the cell cycle at the G0/G1 phase by regulating the expression of p53 and p16. Furthermore, apoptotic morphology, such as chromatin condensation, DNA fragmentation, and apoptotic bodies, was observed in AS-A-treated cells, induced by the activation of the mitochondria-mediated pathway. In an animal study, AS-A reduced tumor volume and prolonged lifespans of mice, with no significant changes in body weight or obvious organ toxicity. This study confirmed the anticancer effects of AS-A by inhibiting cell proliferation, reducing telomerase activity, altering cell cycle progression, and inducing apoptosis. These findings suggest that AS-A has great potential for development as a novel agent or dietary supplement against GB.
Collapse
Affiliation(s)
- Tsung-Liang Ma
- Division of Nephrology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Kai-Fu Chang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Xiao-Fan Huang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Hung-Chih Lai
- Division of Hematology and Oncology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital; Institute of Pharmacology, National Taiwan University, Taipei, Taiwan
| | - Chih-Yen Hsiao
- Division of Nephrology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Nu-Man Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University; Clinical Laboratory, Chung Shan Medical University Hospital, Taichung; Department of Life-and-Death Studies, Nanhua University, Chiayi, Taiwan
| |
Collapse
|
6
|
Effects of Combined Pentadecanoic Acid and Tamoxifen Treatment on Tamoxifen Resistance in MCF−7/SC Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms231911340. [PMID: 36232636 PMCID: PMC9570034 DOI: 10.3390/ijms231911340] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/10/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Estrogen receptors are indicators of breast cancer adaptability to endocrine therapies, such as tamoxifen. Deficiency or absence of estrogen receptor α (ER−α) in breast cancer cells results in reduced efficacy of endocrine therapy. Here, we investigated the effect of combined tamoxifen and pentadecanoic acid therapy on ER−α−under−expressing breast cancer cells. Drug resistance gene expression patterns were determined by RNA sequencing analysis and in vitro experiments. For the first time, we demonstrate that the combined treatment of pentadecanoic acid, an odd−chain fatty acid, and tamoxifen synergistically suppresses the growth of human breast carcinoma MCF−7 stem cells (MCF−7/SCs), which were found to be tamoxifen−resistant and showed reduced ER−α expression compared with the parental MCF−7 cells. In addition, the combined treatment synergistically induced apoptosis and accumulation of sub−G1 cells and suppressed epithelial−to−mesenchymal transition (EMT). Exposure to this combination induces re−expression of ER−α at the transcriptional and protein levels, along with suppression of critical survival signal pathways, such as ERK1/2, MAPK, EGFR, and mTOR. Collectively, decreased ER−α expression was restored by pentadecanoic acid treatment, resulting in reversal of tamoxifen resistance. Overall, pentadecanoic acid exhibits the potential to enhance the efficacy of endocrine therapy in the treatment of ER−α−under−expressing breast cancer cells.
Collapse
|
7
|
Sukocheva OA, Lukina E, Friedemann M, Menschikowski M, Hagelgans A, Aliev G. The crucial role of epigenetic regulation in breast cancer anti-estrogen resistance: Current findings and future perspectives. Semin Cancer Biol 2022; 82:35-59. [PMID: 33301860 DOI: 10.1016/j.semcancer.2020.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/22/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Breast cancer (BC) cell de-sensitization to Tamoxifen (TAM) or other selective estrogen receptor (ER) modulators (SERM) is a complex process associated with BC heterogeneity and the transformation of ER signalling. The most influential resistance-related mechanisms include modifications in ER expression and gene regulation patterns. During TAM/SERM treatment, epigenetic mechanisms can effectively silence ER expression and facilitate the development of endocrine resistance. ER status is efficiently regulated by specific epigenetic tools including hypermethylation of CpG islands within ER promoters, increased histone deacetylase activity in the ER promoter, and/or translational repression by miRNAs. Over-methylation of the ER α gene (ESR1) promoter by DNA methyltransferases was associated with poor prognosis and indicated the development of resistance. Moreover, BC progression and spreading were marked by transformed chromatin remodelling, post-translational histone modifications, and expression of specific miRNAs and/or long non-coding RNAs. Therefore, targeted inhibition of histone acetyltransferases (e.g. MYST3), deacetylases (e.g. HDAC1), and/or demethylases (e.g. lysine-specific demethylase LSD1) was shown to recover and increase BC sensitivity to anti-estrogens. Indicated as a powerful molecular instrument, the administration of epigenetic drugs can regain ER expression along with the activation of tumour suppressor genes, which can in turn prevent selection of resistant cells and cancer stem cell survival. This review examines recent advances in the epigenetic regulation of endocrine drug resistance and evaluates novel anti-resistance strategies. Underlying molecular mechanisms of epigenetic regulation will be discussed, emphasising the utilization of epigenetic enzymes and their inhibitors to re-program irresponsive BCs.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia.
| | - Elena Lukina
- Discipline of Biology, College of Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia
| | - Markus Friedemann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Mario Menschikowski
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Albert Hagelgans
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia; Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432, Russia; Federal State Budgetary Institution «Research Institute of Human Morphology», 3, Tsyurupy Str., Moscow, 117418, Russian Federation; GALLY International Research Institute, San Antonio, TX, 78229, USA.
| |
Collapse
|
8
|
Yen C, Zhao F, Yu Z, Zhu X, Li CG. Interactions Between Natural Products and Tamoxifen in Breast Cancer: A Comprehensive Literature Review. Front Pharmacol 2022; 13:847113. [PMID: 35721162 PMCID: PMC9201062 DOI: 10.3389/fphar.2022.847113] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 04/14/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: Tamoxifen (TAM) is the most commonly used hormone therapeutic drug for the treatment of estrogen receptor-positive (ER+) breast cancer. 30%–70% of clinical breast cancer patients use natural products, which may increase the likelihood of drug interactions. Objective: To evaluate the evidence for the interactions between natural products and TAM in breast cancer. Methods: Electronic databases, including PubMed, CINAHL Plus (via EbscoHost), European PMC, Medline, and Google Scholar, were searched for relevant publications. The search terms include complementary and alternative medicine, natural products, plant products, herbs, interactions, tamoxifen, breast cancer, and their combinations. Results: Various in vitro and in vivo studies demonstrated that the combined use of natural products with TAM produced synergistic anti-cancer effects, including improved inhibition of tumor cell growth and TAM sensitivity and reduced side effects or toxicity of TAM. In contrast, some natural products, including Angelica sinensis (Oliv.) Diels [Apiaceae], Paeonia lactiflora Pall., Rehmannia glutinosa (Gaertn.) DC., Astragalus mongholicus Bunge, and Glycyrrhiza glabra L. [Fabaceae], showed estrogen-like activity, which may reduce the anti-cancer effect of TAM. Some natural products, including morin, silybin, epigallocatechin gallate (EGCG), myricetin, baicalein, curcumin, kaempferol, or quercetin, were found to increase the bioavailability of TAM and its metabolites in vivo. However, three are limited clinical studies on the combination of natural products and TAM. Conclusion: There is evidence for potential interactions of various natural products with TAM in pre-clinical studies, although the relevant clinical evidence is still lacking. Further studies are warranted to evaluate the potential interactions of natural products with TAM in clinical settings.
Collapse
Affiliation(s)
- Christine Yen
- Chinese Medicine Centre, Western Sydney University, Sydney, NSW, Australia.,School of Health Sciences, Western Sydney University, Sydney, NSW, Australia
| | - Fan Zhao
- Chinese Medicine Centre, Western Sydney University, Sydney, NSW, Australia.,College of Chinese Medicine, College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhichao Yu
- Chinese Medicine Centre, Western Sydney University, Sydney, NSW, Australia.,College of the First Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoshu Zhu
- Chinese Medicine Centre, Western Sydney University, Sydney, NSW, Australia.,School of Health Sciences, Western Sydney University, Sydney, NSW, Australia.,NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| | - Chun Guang Li
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| |
Collapse
|
9
|
Hsu RJ, Peng KY, Hsu WL, Chen YT, Liu DW. Z-Ligustilide Induces c-Myc-Dependent Apoptosis via Activation of ER-Stress Signaling in Hypoxic Oral Cancer Cells. Front Oncol 2022; 12:824043. [PMID: 35494068 PMCID: PMC9043595 DOI: 10.3389/fonc.2022.824043] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/10/2022] [Indexed: 12/13/2022] Open
Abstract
Z-ligustilide (or ligustilide) is found in Angelica sinensis (Oliv.) Diels and may exert potential benefits in cancer treatment. Previous research has reported that ligustilide has anti-cancer effects on several types of cancer cells. However, studies of ligustilide on oral cancer cells have not been reported, especially under hypoxic conditions. This study focuses on the molecular mechanism of ligustilide-induced apoptosis in hypoxic oral cancer cells. We found that in hypoxic TW2.6 cells, ligustilide inhibited cell migration and induced caspase-dependent apoptosis. Accumulation of c-Myc accompanied by BH3-only members suggests that ligustilide may induce c-Myc-dependent apoptosis. In addition, we reported that ligustilide has an effect on ER-stress signaling. By using inhibitors of c-Myc, IRE1α, and ER-stress inhibitors, we found that cell morphologies or cell viability were rescued to some degree. Moreover, ligustilide is able to increase the expression of γ-H2AX and enhance the occurrence of DNA damage in oral cancer cells after radiation treatment. This result suggests that ligustilide has potential as a radiation sensitizer. Altogether, we propose that ligustilide may induce c-Myc-dependent apoptosis via ER-stress signaling in hypoxic oral cancer cells.
Collapse
Affiliation(s)
- Ren-Jun Hsu
- Cancer Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Kui-Yuan Peng
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Wen-Lin Hsu
- Cancer Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yu-Tang Chen
- Cancer Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Dai-Wei Liu
- Cancer Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| |
Collapse
|
10
|
Natural Bioactive Compounds Targeting Histone Deacetylases in Human Cancers: Recent Updates. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27082568. [PMID: 35458763 PMCID: PMC9027183 DOI: 10.3390/molecules27082568] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/13/2022]
Abstract
Cancer is a complex pathology that causes a large number of deaths worldwide. Several risk factors are involved in tumor transformation, including epigenetic factors. These factors are a set of changes that do not affect the DNA sequence, while modifying the gene’s expression. Histone modification is an essential mark in maintaining cellular memory and, therefore, loss of this mark can lead to tumor transformation. As these epigenetic changes are reversible, the use of molecules that can restore the functions of the enzymes responsible for the changes is therapeutically necessary. Natural molecules, mainly those isolated from medicinal plants, have demonstrated significant inhibitory properties against enzymes related to histone modifications, particularly histone deacetylases (HDACs). Flavonoids, terpenoids, phenolic acids, and alkaloids exert significant inhibitory effects against HDAC and exhibit promising epi-drug properties. This suggests that epi-drugs against HDAC could prevent and treat various human cancers. Accordingly, the present study aimed to evaluate the pharmacodynamic action of different natural compounds extracted from medicinal plants against the enzymatic activity of HDAC.
Collapse
|
11
|
Natural Bioactive Compounds Targeting Epigenetic Pathways in Cancer: A Review on Alkaloids, Terpenoids, Quinones, and Isothiocyanates. Nutrients 2021; 13:nu13113714. [PMID: 34835969 PMCID: PMC8621755 DOI: 10.3390/nu13113714] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is one of the most complex and systemic diseases affecting the health of mankind, causing major deaths with a significant increase. This pathology is caused by several risk factors, of which genetic disturbances constitute the major elements, which not only initiate tumor transformation but also epigenetic disturbances which are linked to it and which can induce transcriptional instability. Indeed, the involvement of epigenetic disturbances in cancer has been the subject of correlations today, in addition to the use of drugs that operate specifically on different epigenetic pathways. Natural molecules, especially those isolated from medicinal plants, have shown anticancer effects linked to mechanisms of action. The objective of this review is to explore the anticancer effects of alkaloids, terpenoids, quinones, and isothiocyanates.
Collapse
|
12
|
Yang J, Xing Z. Ligustilide counteracts carcinogenesis and hepatocellular carcinoma cell-evoked macrophage M2 polarization by regulating yes-associated protein-mediated interleukin-6 secretion. Exp Biol Med (Maywood) 2021; 246:1928-1937. [PMID: 34053234 DOI: 10.1177/15353702211010420] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cross-communication between cancer cells and macrophages within the tumor microenvironment fulfills the critical roles in the progression of cancers, including hepatocellular carcinoma (HCC). Ligustilide exerts anti-inflammation, anti-injury, and anti-tumor pleiotropic pharmacological functions. Nevertheless, its roles in HCC cells and tumor microenvironment remain elusive. In the current study, ligustilide dramatically restrained HCC cell viability and migration but had little cytotoxicity to normal hepatocytes. Importantly, ligustilide antagonized HCC cell co-culture-induced macrophage recruitment and M2 polarization by enhancing the percentage of CD14+CD206+ cells and macrophage M2 markers (CD163, Arg1, CD206, CCL22, IL-10, and TGF-β). Mechanistically, ligustilide repressed yes-associated protein (YAP) activation by reducing nuclear translocation, protein expression, transcriptional regulatory activity of YAP, and increasing p-YAP levels. Noticeably, blocking the YAP offset the suppressive effects of ligustilide on macrophage recruitment and M2 polarization evoked by HCC cells. Moreover, the release of interleukin-6 (IL-6) was mitigated by ligustilide in a YAP-dependent manner in HCC cells, concomitant with inhibition of IL-6R/STAT3 signaling activation. Of interest, interdicting the IL-6 aggravated ligustilide-mediated suppression in HCC-induced macrophage recruitment and M2 polarization; whereas exogenous IL-6 treatment reversed the above effects. Additionally, blockage of IL-6R signaling also overturned IL-6-induced macrophage recruitment and M2 phenotype. Consequently, these findings support a notion that ligustilide not only restrains HCC cell malignancy but also antagonizes HCC cell-evoked macrophage recruitment and M2 polarization by inhibiting YAP/IL-6 release-induced activation of the IL-6 receptor/signal transducer and activator of transcription 3 (IL-6R/STAT3) signaling. Thus, ligustilide may be a promising therapeutic agent to fight HCC by regulating cancer cells and cross-talk between tumor cells and macrophages in tumor microenvironment.
Collapse
Affiliation(s)
- Jikang Yang
- Department of Gastroenterology, Jiaozuo People's Hospital, Jiaozuo 454000, China
| | - Zhiyuan Xing
- Emergency Department, Jiaozuo Hospital of Traditional Chinese Medicine, Jiaozuo 454150, China
| |
Collapse
|
13
|
Chao J, Ko CY, Lin CY, Tomoji M, Huang CH, Chiang HC, Yang JJ, Huang SS, Su SY. Ethnobotanical Survey of Natural Galactagogues Prescribed in Traditional Chinese Medicine Pharmacies in Taiwan. Front Pharmacol 2021; 11:625869. [PMID: 33679390 PMCID: PMC7928277 DOI: 10.3389/fphar.2020.625869] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/23/2020] [Indexed: 01/14/2023] Open
Abstract
Natural medicinal materials have been used to promote breast milk secretion. Here, we investigated the natural medicinal materials prescribed in traditional Chinese medicine (TCM) pharmacies across Taiwan to induce lactation. We collected medicinal materials from 87 TCM pharmacies, identified them in the prescriptions, and analyzed their drug contents. We examined their botanical origins, biological classifications, traditional usage, and modern pharmacological properties. We used the TCM Inheritance Support System to identify core medicinal materials in galactogenous prescriptions. We collected 81 medicinal materials from 90 galactogenous prescriptions. Leguminosae accounted for 12%, whereas Apiaceae accounted for 7% of all materials examined. The primary medicinal plant parts used were roots and seeds. Nineteen frequently used medicinal materials had a relative frequency of citation of greater than or equal to 0.2. According to their efficacy, 58% were warm, 54% were sweet, and 63% were tonifying; 74% of the frequently used medicinal materials have been showed efficacy against breast cancer. The primary core medicinal material was Angelica sinensis (Oliv.) Diels, whereas the secondary core medicinal materials were Tetrapanax papyrifer (Hook.) K. Koch and Hedysarum polybotrys Hand.-Mazz. Most galactogenous prescriptions consisted of multiple materials from Leguminosae and Apiaceae. The mechanisms underlying galactogenous efficacy warrant further investigations.
Collapse
Affiliation(s)
- Jung Chao
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Chien-Yu Ko
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Chin-Yu Lin
- Institute of New Drug Development, China Medical University, Taichung, Taiwan.,Tsuzuki Institute for Traditional Medicine, China Medical University, Taichung, Taiwan
| | - Maeda Tomoji
- Department of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama, Japan.,Tsuzuki Institute for Traditional Medicine, China Medical University, Taichung, Taiwan
| | | | - Hung-Che Chiang
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Jeng-Jer Yang
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Shyh-Shyun Huang
- School of Pharmacy, China Medical University, Taichung, Taiwan.,Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan
| | - Shan-Yu Su
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan.,School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
14
|
Zhang B, Wu D, Hu L, Cha X, Liu Y, Li J, Xie B, Zheng L, Li B. Ligustilide inhibits the proliferation of human osteoblastoma MG63 cells through the TLR4-ERK pathway. Life Sci 2021; 288:118993. [PMID: 33545202 DOI: 10.1016/j.lfs.2020.118993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/20/2020] [Accepted: 12/24/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To study the proapoptotic effect of ligustilide on osteoblastoma (OS) and the relative related molecular mechanism. METHODS AND MATERIALS An MTT was used to examine the proliferation of OS cells, and Flow cytometry was used to analyze apoptosis and the cell cycle. Western blotting was used to detect the signaling pathway of apoptosis, and immunohistochemical (IH) staining was used to detect the apoptosis status of OS cells. A TLR4 inhibitor was used to study the effect of ligustilide on OS. RESULTS Ligustilide inhibited OS cell proliferation but had no inhibitory effect on normal bone marrow cells. Flow cytometry results showed that ligustilide induced apoptosis in OS cells, and the cell cycle was arrested at the M/G2 phase. Western blot results showed that ERK, P53, P21, Caspase 9, Caspase 8 and Caspase 3 were all activated; cytochrome C and Bax increased; and Bcl-2 decreased when OS was treated with ligustilide. When an ERK or Caspase inhibitor was added to the culture medium, the apoptosis of OS cells decreased to some degree. When OS cells were pretreated with CLI-095, which is a TLR4 inhibitor, the percentage of apoptotic cells and cell cycle arrest were both reversed. IH results also showed that ligustilide induced apoptosis in OS cells, and the effect was blocked by the TLR4 inhibitor. CONCLUSION Ligustilide selectively inhibited the proliferation of OS cells by inducing apoptosis, which possibly included endogenous and exogenous apoptosis through TLR4.
Collapse
Affiliation(s)
- Bin Zhang
- Luzhou City Hospital of Traditional Chinese Medicine, Luzhou, China
| | - Donghai Wu
- Luzhou City Hospital of Traditional Chinese Medicine, Luzhou, China
| | - Limei Hu
- Luzhou City Hospital of Traditional Chinese Medicine, Luzhou, China
| | - Xiaofeng Cha
- Luzhou City Hospital of Traditional Chinese Medicine, Luzhou, China
| | - Yilai Liu
- Luzhou City Hospital of Traditional Chinese Medicine, Luzhou, China
| | - Jujie Li
- Luzhou City Hospital of Traditional Chinese Medicine, Luzhou, China
| | - Bo Xie
- Luzhou City Hospital of Traditional Chinese Medicine, Luzhou, China
| | - Lei Zheng
- Luzhou City Hospital of Traditional Chinese Medicine, Luzhou, China..
| | - Bin Li
- Luzhou City Hospital of Traditional Chinese Medicine, Luzhou, China..
| |
Collapse
|
15
|
Wang C, Liu G, Dou G, Yang Y, Chen L, Ma H, Jiang Z, Ma H, Li C, Li L, Jiang M, Lu Q, Li P, Qi H. Z-Ligustilide Selectively Targets AML by Restoring Nuclear Receptors Nur77 and NOR-1-mediated Apoptosis and Differentiation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 82:153448. [PMID: 33421904 DOI: 10.1016/j.phymed.2020.153448] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 11/12/2020] [Accepted: 12/22/2020] [Indexed: 05/27/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a devastating hematologic malignancy with a high mortality. The nuclear receptors Nur77 and NOR-1 are commonly downregulated in human AML blasts and have emerged as key therapeutic targets for AML. METHODS This study aimed to identify Z-ligustilide (Z-LIG), the main phthalide of Rhizoma Chuanxiong, as a potential agent that can selectively target AML. The anti-AML activity of Z-LIG was evaluated in vitro and in vivo, and the effect and underlying mechanisms of Z-LIG on the restoration of Nur77 and NOR-1 was determined. Moreover, the role of Nur77 and NOR-1 in the regulation of Z-LIG-induced apoptosis and differentiation of AML cells was explored. RESULTS Z-LIG preferentially inhibited the viability of human AML cells, as well as suppressed the proliferation and colony formation ability. Notably, a concentration-dependent dual effect of Z-LIG was observed in AML cells: inducing apoptosis at relatively high concentrations (25 μM to 100 μM) and promoting differentiation at relatively low concentrations (10 μM and 25 μM). Importantly, Z-LIG restored Nur77 and NOR-1 expression in AML cells by increasing Ace-H3 (lys9/14) enrichment in their promoters. Meanwhile, Z-LIG enhanced the recruitment of p300 and reduced the recruitment of HDAC1, HDAC4/5/7, and MTA1 in the Nur77 promoter and enhanced the recruitment of p-CREB and reduced HDAC1 and HDAC3 in the NOR-1 promoter. Furthermore, Z-LIG-induced apoptosis was shown to be correlated with the mitochondria localization of Nur77/NOR-1 and subsequent Bcl-2 conformational change, converting Bcl-2 from a cyto-protective phenotype into a cyto-destructive phenotype. Z-LIG-promoted differentiation was found to be related to Nur77/NOR-1-mediated myeloid differentiation-associated transcription factors Jun B, c-Jun, and C/EBPβ. Finally, silencing of Nur77 and NOR-1 attenuated anti-AML activity of Z-LIG in NOD/SCID mice. CONCLUSIONS Our study suggests that Z-LIG may serve as a novel bifunctional agent for AML by restoring Nur77/NOR-1-mediated apoptosis and differentiation.
Collapse
Affiliation(s)
- Chengqiang Wang
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Gen Liu
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Guojun Dou
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Yi Yang
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Lu Chen
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Hui Ma
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Zhuyun Jiang
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Haoyue Ma
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Chenglong Li
- Department of Hematology, Sichuan Provincial People's Hospital, Chengdu 610212, Sichuan, China
| | - Li Li
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Mingdong Jiang
- Department of Oncology and Hematology, Chongqing Ninth People's Hospital, Jialing Village 69, Beibei District, Chongqing 400700, China
| | - Qianwei Lu
- Department of Oncology and Hematology, Chongqing Ninth People's Hospital, Jialing Village 69, Beibei District, Chongqing 400700, China
| | - Pan Li
- Department of Oncology and Hematology, Chongqing Ninth People's Hospital, Jialing Village 69, Beibei District, Chongqing 400700, China
| | - Hongyi Qi
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China.
| |
Collapse
|
16
|
Jiang X, Zhao W, Zhu F, Wu H, Ding X, Bai J, Zhang X, Qian M. Ligustilide inhibits the proliferation of non-small cell lung cancer via glycolytic metabolism. Toxicol Appl Pharmacol 2020; 410:115336. [PMID: 33212065 DOI: 10.1016/j.taap.2020.115336] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/30/2020] [Accepted: 11/12/2020] [Indexed: 12/20/2022]
Abstract
Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer-related death worldwide. The abnormal activation of glycolytic metabolism and PTEN/AKT signaling in NSCLC cells are highly correlated with their proliferation abilities and viability. Ligustilide is one of the major bioactive components of multiple Chinese traditional medicine including Angelica sinensis and Ligusticum. Ligustilide exposure inhibits the proliferation and viability of multiple cancer cell lines in vitro. However, the impact of ligustilide to the progression of NSCLC and its detailed pharmacological mechanisms remain unclear. In this research, CCK-8 and colony formation assay were performed to demonstrate ligustilide treatment inhibited the viability and proliferation ability of NSCLC cells in vitro. Caspase-3/-7 activity assay and nucleosome ELISA assay were utilized to show ligustilide promoted the apoptosis of NSCLC cells. Metabolic analysis and qRT-PCR assay were used to demonstrated that ligustilide dampened aerobic glycolysis of NSCLC cells. Nude mice were exposed to 5 mg/kg ligustilide and ligustilide inhibited orthotopic NSCLC growth in vivo. qRT-PCR and Western blot analysis were performed to substantiate the regulatory function of ligustilide to PTEN/AKT signaling in NSCLC cells. Overall, this study revealed that ligustilide regulated the proliferation, apoptosis and aerobic glycolysis of NSCLC cells through PTEN/AKT signaling pathway.
Collapse
Affiliation(s)
- Xiufeng Jiang
- Wuxi Fifth People's Hospital, Wuxi 214016, Jiangsu, China.
| | - Wei Zhao
- Wuxi Fifth People's Hospital, Wuxi 214016, Jiangsu, China
| | - Feng Zhu
- Wuxi Fifth People's Hospital, Wuxi 214016, Jiangsu, China
| | - Hui Wu
- Wuxi Fifth People's Hospital, Wuxi 214016, Jiangsu, China
| | - Xiao Ding
- Wuxi Fifth People's Hospital, Wuxi 214016, Jiangsu, China
| | - Jinmei Bai
- Wuxi Fifth People's Hospital, Wuxi 214016, Jiangsu, China
| | - Xiaoqing Zhang
- Wuxi Fifth People's Hospital, Wuxi 214016, Jiangsu, China
| | - Meifang Qian
- Wuxi Fifth People's Hospital, Wuxi 214016, Jiangsu, China
| |
Collapse
|
17
|
Akone SH, Ntie-Kang F, Stuhldreier F, Ewonkem MB, Noah AM, Mouelle SEM, Müller R. Natural Products Impacting DNA Methyltransferases and Histone Deacetylases. Front Pharmacol 2020; 11:992. [PMID: 32903500 PMCID: PMC7438611 DOI: 10.3389/fphar.2020.00992] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 06/19/2020] [Indexed: 12/24/2022] Open
Abstract
Epigenetics refers to heritable changes in gene expression and chromatin structure without change in a DNA sequence. Several epigenetic modifications and respective regulators have been reported. These include DNA methylation, chromatin remodeling, histone post-translational modifications, and non-coding RNAs. Emerging evidence has revealed that epigenetic dysregulations are involved in a wide range of diseases including cancers. Therefore, the reversible nature of epigenetic modifications concerning activation or inhibition of enzymes involved could be promising targets and useful tools for the elucidation of cellular and biological phenomena. In this review, emphasis is laid on natural products that inhibit DNA methyltransferases (DNMTs) and histone deacetylases (HDACs) making them promising candidates for the development of lead structures for anticancer-drugs targeting epigenetic modifications. However, most of the natural products targeting HDAC and/or DNMT lack isoform selectivity, which is important for determining their potential use as therapeutic agents. Nevertheless, the structures presented in this review offer the well-founded basis that screening and chemical modifications of natural products will in future provide not only leads to the identification of more specific inhibitors with fewer side effects, but also important features for the elucidation of HDAC and DNMT function with respect to cancer treatment.
Collapse
Affiliation(s)
- Sergi Herve Akone
- Department of Chemistry, Faculty of Science, University of Douala, Douala, Cameroon
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research and Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Fidele Ntie-Kang
- Department of Chemistry, Faculty of Science, University of Buea, Buea, Cameroon
- Institute for Pharmacy, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
- Institut für Botanik, Technische Universität Dresden, Dresden, Germany
| | - Fabian Stuhldreier
- Medical Faculty, Institute of Molecular Medicine I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Alexandre Mboene Noah
- Department of Biochemistry, Faculty of Science, University of Douala, Douala, Cameroon
| | | | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research and Department of Pharmacy, Saarland University, Saarbrücken, Germany
| |
Collapse
|
18
|
Xie Q, Zhang L, Xie L, Zheng Y, Liu K, Tang H, Liao Y, Li X. Z‐ligustilide: A review of its pharmacokinetics and pharmacology. Phytother Res 2020; 34:1966-1991. [DOI: 10.1002/ptr.6662] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/17/2020] [Accepted: 02/16/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Qingxuan Xie
- School of PharmacyChengdu University of Traditional Chinese Medicine Chengdu China
| | - Linlin Zhang
- School of PharmacyChengdu University of Traditional Chinese Medicine Chengdu China
| | - Long Xie
- School of PharmacyChengdu University of Traditional Chinese Medicine Chengdu China
| | - Yu Zheng
- School of PharmacyChengdu University of Traditional Chinese Medicine Chengdu China
| | - Kai Liu
- School of PharmacyChengdu University of Traditional Chinese Medicine Chengdu China
| | - Hailong Tang
- School of PharmacyChengdu University of Traditional Chinese Medicine Chengdu China
| | - Yanmei Liao
- School of PharmacyChengdu University of Traditional Chinese Medicine Chengdu China
| | - Xiaofang Li
- School of PharmacyChengdu University of Traditional Chinese Medicine Chengdu China
| |
Collapse
|
19
|
Ma J, Mei J, Lu J, Wang Y, Hu M, Ma F, Long H, Qin Z, Tao N. Ligustilide promotes apoptosis of cancer-associated fibroblasts via the TLR4 pathways. Food Chem Toxicol 2020; 135:110991. [DOI: 10.1016/j.fct.2019.110991] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 10/19/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022]
|
20
|
Chen JY, Wang YH, Hidajah AC, Li CY. A population-based case-control study on the association of Angelica sinensis exposure with risk of breast cancer. J Tradit Complement Med 2019; 10:454-459. [PMID: 32953561 PMCID: PMC7484959 DOI: 10.1016/j.jtcme.2019.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 10/15/2019] [Accepted: 10/19/2019] [Indexed: 01/09/2023] Open
Abstract
Background Due to a lack of evidence from large-scale epidemiological studies by far on this issue, whether there is a link between Angelica sinensis exposure and breast cancer risk remained inconclusive. Methods We conducted a population-based case-control study using Taiwan’s National Health Insurance claim data, in which all breast cancer patients newly diagnosed between 2005 and 2008 were employed as the case group (n = 34,262) and a random sample of non-breast cancer individuals selected from 1-million beneficiaries registered in 2005 was served as the control group. For fair comparability, we employed the time density sampling method to select controls who were matched to case on date of breast cancer diagnosis and age with a case/control ratio of 1/3 (n = 102,786). Results We found that the use of Angelica sinensis presents a weakly but significantly protective effect on breast cancer (adjusted odds ratio (aOR) 0.95, 95% confidence interval (CI) 0.93–0.98), with a significant dose-gradient relationship. We also noted a stronger association with breast cancer with initial use of Angelica sinensis at a longer time before breast cancer diagnosis, and found that the seemingly protective effect of Angelica sinensis was more obvious among women who had initial use at 47–55 years (aOR 0.93, 95% CI 0.88–0.98). Conclusion This population-based case-control study revealed that exposure to Angelica sinensis showed a weakly but significantly protective effect on breast cancer risk, which could ease people’s concern over the potential carcinogenic effect from exposure to Angelica sinensis. Angelica sinensis was associated with a weakly protective effect on breast cancer. The methodological strengths included population-based and a fairy large sample size. Provide further insight to the link between phytohormone in TCM and breast cancer.
Collapse
Affiliation(s)
- Jhong-Yuan Chen
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Kaohsiung, Taiwan.,Department of Public Health, College of Medicine, National Cheng Kung University, No.1, University Road, East District, Tainan, Taiwan
| | - Yi-Hsiu Wang
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Kaohsiung, Taiwan
| | - Atik Choirul Hidajah
- Department of Epidemiology, Faculty of Public Health, Universitas Airlangga, Kampus C UNAIR, Jl. Mulyorejo Surabaya, East Java, Indonesia
| | - Chung-Yi Li
- Department of Public Health, College of Medicine, National Cheng Kung University, No.1, University Road, East District, Tainan, Taiwan.,Department of Epidemiology, Faculty of Public Health, Universitas Airlangga, Kampus C UNAIR, Jl. Mulyorejo Surabaya, East Java, Indonesia.,Department of Public Health, College of Public Health, China Medical University, No.91, Xueshi Road, North District, Taichung, Taiwan
| |
Collapse
|
21
|
Yue GGL, Wong LS, Leung HW, Gao S, Tsang JYS, Lin ZX, Law BKB, Tse GMK, Lau CBS. Is Danggui Safe to be Taken by Breast Cancer Patients?-A Skepticism Finally Answered by Comprehensive Preclinical Evidence. Front Pharmacol 2019; 10:706. [PMID: 31293425 PMCID: PMC6604035 DOI: 10.3389/fphar.2019.00706] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/31/2019] [Indexed: 11/23/2022] Open
Abstract
Angelica sinensis (AS, Danggui) has long been regarded to stimulate breast cancer growth; hence, the use of AS in breast cancer patients remains a major concern for both patients and practitioners. Since safety studies of herbs would be unethical to carry out in patients, the present study aimed to investigate the potential unsafe effects of AS in a systematic pre-clinical approach. Human breast cancer cells, breast orthotopic tumor-bearing mouse models, as well as primary breast cancer cells from patients’ tumors were used to evaluate the effect of AS hot water extract on the progression of breast tumors and/or growth of breast cancer cells. We showed that AS is not that stimulatory in breast cancer both in vitro and in vivo, though AS should still be used with caution in estrogen receptor-positive breast cancer patients. This novel approach of applying breast cancer cell lines, xenograft, and syngeneic tumors models, as well as primary breast cancer cells from patients’ tumors in Chinese medicines safety evaluation was proven feasible. Our finding is important information for patients, Chinese medicine practitioners, and clinicians on the safety use of AS in breast cancer, which will affect future clinical practice.
Collapse
Affiliation(s)
- Grace Gar-Lee Yue
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Lok-Sze Wong
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Hoi-Wing Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Si Gao
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Julia Yuen-Shan Tsang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Zhi-Xiu Lin
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | | - Gary Man-Kit Tse
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Clara Bik-San Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| |
Collapse
|
22
|
Ma J, Xu Y, Zheng Q, Wang Y, Hu M, Ma F, Long H, Qin Z, Tao N. Ligustilide inhibits the activation of cancer-associated fibroblasts. Life Sci 2019; 218:58-64. [DOI: 10.1016/j.lfs.2018.12.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/16/2018] [Accepted: 12/18/2018] [Indexed: 11/27/2022]
|
23
|
Apoptotic cell death induced by Z-Ligustilidein human ovarian cancer cells and role of NRF2. Food Chem Toxicol 2018; 121:631-638. [DOI: 10.1016/j.fct.2018.09.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/06/2018] [Accepted: 09/19/2018] [Indexed: 02/07/2023]
|
24
|
Abstract
Macroautophagy/autophagy is vital for intracellular quality control and homeostasis. Therefore, careful regulation of autophagy is very important. In the past 10 years, a number of studies have reported that estrogenic effectors affect autophagy. However, some results, especially those regarding the modulatory effect of 17β-estradiol (E2) on autophagy seem inconsistent. Moreover, several clinical trials are already in place combining both autophagy inducers and autophagy inhibitors with endocrine therapies for breast cancer. Not all patients experience benefit, which further confuses and complicates our understanding of the main effects of autophagy in estrogen-related cancer. In view of the importance of the crosstalk between estrogen signaling and autophagy, this review summarizes the estrogenic effectors reported to affect autophagy, subcellular distribution and translocation of estrogen receptors, autophagy-targeted transcription factors (TFs), miRNAs, and histone modifications regulated by E2. Upon stimulation with estrogen, there will always be opposing functional actions, which might occur between different receptors, receptors on TFs, TFs on autophagy genes, or even histone modifications on transcription. The huge signaling network downstream of estrogen can promote autophagy and reduce overstimulated autophagy at the same time, which allows autophagy to be regulated by estrogen in a restricted range. To help understand how the estrogenic regulation of autophagy affects cell fate, a hypothetical model is presented here. Finally, we discuss some exciting new directions in the field. We hope this might help to better understand the multiple associations between estrogen and autophagy, the pathogenic mechanisms of many estrogen-related diseases, and to design novel and efficacious therapeutics. Abbreviations: AP-1, activator protein-1; HATs, histone acetyltransferases; HDAC, histone deacetylases; HOTAIR, HOX transcript antisense RNA.
Collapse
Affiliation(s)
- Jin Xiang
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Xiang Liu
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Jing Ren
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Kun Chen
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Hong-Lu Wang
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Yu-Yang Miao
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Miao-Miao Qi
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| |
Collapse
|
25
|
Co-inhibition of mTORC1, HDAC and ESR1α retards the growth of triple-negative breast cancer and suppresses cancer stem cells. Cell Death Dis 2018; 9:815. [PMID: 30050079 PMCID: PMC6062597 DOI: 10.1038/s41419-018-0811-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most refractory subtype of breast cancer. It causes the majority of breast cancer-related deaths, which has been largely associated with the plasticity of tumor cells and persistence of cancer stem cells (CSCs). Conventional chemotherapeutics enrich CSCs and lead to drug resistance and disease relapse. Development of a strategy capable of inhibiting both bulk and CSC populations is an unmet medical need. Inhibitors against estrogen receptor 1, HDACs, or mTOR have been studied in the treatment of TNBC; however, the results are inconsistent. In this work, we found that patient TNBC samples expressed high levels of mTORC1 and HDAC genes in comparison to luminal breast cancer samples. Furthermore, co-inhibition of mTORC1 and HDAC with rapamycin and valproic acid, but neither alone, reproducibly promoted ESR1 expression in TNBC cells. In combination with tamoxifen (inhibiting ESR1), both S6RP phosphorylation and rapamycin-induced 4E-BP1 upregulation in TNBC bulk cells was inhibited. We further showed that fractionated CSCs expressed higher levels of mTORC1 and HDAC than non-CSCs. As a result, co-inhibition of mTORC1, HDAC, and ESR1 was capable of reducing both bulk and CSC subpopulations as well as the conversion of fractionated non-CSC to CSCs in TNBC cells. These observations were partially recapitulated with the cultured tumor fragments from TNBC patients. Furthermore, co-administration of rapamycin, valproic acid, and tamoxifen retarded tumor growth and reduced CD44high/+/CD24low/− CSCs in a human TNBC xenograft model and hampered tumorigenesis after secondary transplantation. Since the drugs tested are commonly used in clinic, this study provides a new therapeutic strategy and a strong rationale for clinical evaluation of these combinations for the treatment of patients with TNBC.
Collapse
|
26
|
Zhang Y, Wang J, Zuo C, Chen W, Zhu Q, Guo D, Wu H, Wang H, Peng D, Han L. Protective Effect of Taohong Siwu Decoction on Abnormal Uterine Bleeding Induced by Incomplete Medical Abortion in Rats during Early Pregnancy. Chem Pharm Bull (Tokyo) 2018; 66:708-713. [DOI: 10.1248/cpb.c17-00945] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yanyan Zhang
- School of Pharmacy, Anhui University of Chinese Medicine
- Department of Pharmacy, Anhui Medical College
- Anhui Provincial Key Laboratory for Chinese Medicine Research and Development
| | - Jichen Wang
- School of Pharmacy, Anhui University of Chinese Medicine
- Anhui Provincial Key Laboratory for Chinese Medicine Research and Development
| | - Chijing Zuo
- School of Pharmacy, Anhui University of Chinese Medicine
- Anhui Provincial Key Laboratory for Chinese Medicine Research and Development
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine
- Anhui Provincial Key Laboratory for Chinese Medicine Research and Development
| | - Qian Zhu
- Anhui Key Laboratory of Bioactivity of Natural Products
- School of Pharmacy, Institute for Liver Diseases of Anhui Medical University
| | - Dongdong Guo
- School of Pharmacy, Anhui University of Chinese Medicine
- Anhui Provincial Key Laboratory for Chinese Medicine Research and Development
| | - Huanru Wu
- School of Pharmacy, Anhui University of Chinese Medicine
- Anhui Provincial Key Laboratory for Chinese Medicine Research and Development
| | - Huizhuo Wang
- School of Pharmacy, Anhui University of Chinese Medicine
- Anhui Provincial Key Laboratory for Chinese Medicine Research and Development
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine
- Anhui Provincial Key Laboratory for Chinese Medicine Research and Development
| | - Lan Han
- School of Pharmacy, Anhui University of Chinese Medicine
- Anhui Provincial Key Laboratory for Chinese Medicine Research and Development
| |
Collapse
|
27
|
Zhao Y, Liu Y. A mechanistic overview of herbal medicine and botanical compounds to target transcriptional factors in Breast cancer. Pharmacol Res 2017; 130:292-302. [PMID: 29292214 DOI: 10.1016/j.phrs.2017.12.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 11/23/2017] [Accepted: 12/23/2017] [Indexed: 12/28/2022]
Abstract
The abnormalities of transcription factors, such as NF-κB, STAT, estrogen receptor, play a critical role in the initiation and progression of breast cancer. Due to the limitation of current treatment, transcription factors could be promising therapeutic targets, which have received close attention. In this review, we introduced herbal medicines, as well as botanical compounds that had been verified with anti-tumor properties via regulating transcription factors. Herbs, compounds, as well as formulae reported with various transcriptional targets, were summarized thoroughly, to provide implication for the future research on basic experiment and clinical application.
Collapse
Affiliation(s)
- Yingke Zhao
- Cardiovascular Diseases Centre, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| | - Yue Liu
- Cardiovascular Diseases Centre, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
28
|
Antiangiogenic activity of phthalides-enriched Angelica Sinensis extract by suppressing WSB-1/pVHL/HIF-1α/VEGF signaling in bladder cancer. Sci Rep 2017; 7:5376. [PMID: 28710377 PMCID: PMC5511260 DOI: 10.1038/s41598-017-05512-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 05/31/2017] [Indexed: 12/28/2022] Open
Abstract
The hypoxia-inducible factor-1α (HIF-1α) plays a critical role in tumor angiogenesis. It has been reported that the acetone extract of Angelica sinensis (AE-AS) rich in phthalides is able to inhibit cancer cell proliferation. However, whether AE-AS reduces cancer angiogenesis remains unknown. In this study, we demonstrated that AE-AS significantly inhibited the angiogenesis in vitro and in vivo evidenced by attenuation of the tube formation in hypoxic human umbilical vascular endothelial cells (HUVECs), and the vasculature generation in Matrigel plug, the chicken chorioallantoic membrane, and tumors. Treatment with AE-AS markedly decreased the protein accumulation and transcriptional activity of HIF-1α, vascular endothelial growth factor (VEGF) expression/secretion, and VEGFR2 phosphorylation in hypoxic human bladder cancer (T24) cells and tumor tissues accompanied by a reduction of tumor growth. Notably, AE-AS-induced HIF-1α protein degradation may, at least partly, attribute to inhibition of WSB-1-dependent pVHL degradation. Moreover, VEGFR2-activated PI3K/AKT/mTOR signaling pathway in hypoxic T24 cells was greatly inhibited by AE-AS. Collectively, AE-AS may be a potential anticancer agent by attenuating cancer angiogenesis via suppression of WSB-1/pVHL/HIF-1α/VEGF/VEGFR2 cascade.
Collapse
|