1
|
Wei L, Wang B, Bai J, Zhang Y, Liu C, Suo H, Wang C. Postbiotics are a candidate for new functional foods. Food Chem X 2024; 23:101650. [PMID: 39113733 PMCID: PMC11304867 DOI: 10.1016/j.fochx.2024.101650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
Accumulating studies have highlighted the great potential of postbiotics in alleviating diseases and protecting host health. Compared with traditional functional foods (such as probiotics and prebiotics), postbiotics have the advantages of a single composition, high physiological activity, long shelf life, easy absorption, and high targeting, etc. The development of postbiotics has led to a wide range of potential applications in functional food and drug development. However, the lack of clinical trial data, mechanism analyses, safety evaluations, and effective regulatory frameworks has limited the application of postbiotic products. This review describes the definition, classification, sources, and preparation methods of postbiotics, the progress and mechanism of preclinical and clinical research in improving host diseases, and their application in food. Strengthen understanding of the recognition and development of related products to lay a theoretical foundation.
Collapse
Affiliation(s)
- Li Wei
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Botao Wang
- Bloomage Biotechnology CO, LTD, Jinan, Shandong 250000, China
| | - Junying Bai
- Citrus Research Institute, Southwest University, Chongqing 400700, China
| | - Yuyan Zhang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Cuiping Liu
- Department of Radiology, Yuxi Children's Hospital, Yuxi, Yunnan 653100, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China
| |
Collapse
|
2
|
Hosseini MS, Sanaat Z, Akbarzadeh MA, Vaez-Gharamaleki Y, Akbarzadeh M. Histone deacetylase inhibitors for leukemia treatment: current status and future directions. Eur J Med Res 2024; 29:514. [PMID: 39456044 PMCID: PMC11515273 DOI: 10.1186/s40001-024-02108-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Leukemia remains a major therapeutic challenge in clinical oncology. Despite significant advancements in treatment modalities, leukemia remains a significant cause of morbidity and mortality worldwide, as the current conventional therapies are accompanied by life-limiting adverse effects and a high risk of disease relapse. Histone deacetylase inhibitors have emerged as a promising group of antineoplastic agents due to their ability to modulate gene expression epigenetically. In this review, we explore these agents, their mechanisms of action, pharmacokinetics, safety and clinical efficacy, monotherapy and combination therapy strategies, and clinical challenges associated with histone deacetylase inhibitors in leukemia treatment, along with the latest evidence and ongoing studies in the field. In addition, we discuss future directions to optimize the therapeutic potential of these agents.
Collapse
Affiliation(s)
- Mohammad-Salar Hosseini
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 51666, EA, Iran.
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Evidence-Based Medicine, Iranian EBM Center: A JBI Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Zohreh Sanaat
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amin Akbarzadeh
- Research Center for Evidence-Based Medicine, Iranian EBM Center: A JBI Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yosra Vaez-Gharamaleki
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Akbarzadeh
- Research Center for Evidence-Based Medicine, Iranian EBM Center: A JBI Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Ibrahim JN, El-Hakim S, Semaan J, Ghosn S, El Ayoubi H, Elnar AA, Tohme N, El Boustany C. Sodium Butyrate (NaB) and Sodium Propionate (NaP) Reduce Cyclin A2 Expression, Inducing Cell Cycle Arrest and Proliferation Inhibition of Different Breast Cancer Subtypes, Leading to Apoptosis. Biomedicines 2024; 12:1779. [PMID: 39200243 PMCID: PMC11351769 DOI: 10.3390/biomedicines12081779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/27/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Sodium butyrate (NaB) and sodium propionate (NaP) have recently garnered attention for their role in regulating inflammation and controlling signaling pathways of cell growth and apoptosis, potentially preventing cancer development. However, their therapeutic effect and the underlying mechanisms involved remain elusive in breast cancer. This study aims at investigating the anticancer role of NaB and NaP in different types of breast cancer by assessing their antiproliferative effect on MCF-7 and MDA-MB-231 cells (through an MTT assay), as well as their ability to alter the cell cycle and cyclin expression (using flow cytometry and RT-qPCR, respectively), and to promote apoptosis (using Annexin V-FITC conjugated and sub-G1 phase techniques). MDA-MB-231 cell proliferation was inhibited by NaB and NaP in a dose- and time-dependent manner with respective IC50 values of 2.56 mM and 6.49 mM. Treatment induced cell arrest in the G1 phase which was further supported by the significant reduction in cyclin A2 and cyclin B1 expressions. Finally, NaB, and less significantly NaP, induced apoptosis in a dose-dependent manner with higher concentrations required for MDA-MB-231 than MCF-7. Our findings elucidate the cyclin-dependent inhibitory effect of NaB and NaP on the progression of different breast cancer subtypes, thus highlighting their therapeutic potential in breast cancer.
Collapse
Affiliation(s)
- José-Noel Ibrahim
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut 1102, Lebanon
| | - Sandy El-Hakim
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait;
| | - Josiane Semaan
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| | - Stéphanie Ghosn
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| | - Hiba El Ayoubi
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| | - Arpiné Ardzivian Elnar
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| | - Najat Tohme
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| | - Charbel El Boustany
- Department of Laboratory Science, Faculty of Public Health—Branch 2, Lebanese University, Fanar 2611, Lebanon; (J.S.); (S.G.); (H.E.A.); (A.A.E.); (N.T.); (C.E.B.)
| |
Collapse
|
4
|
Soldado-Gordillo A, Álvarez-Mercado AI. Epigenetics, Microbiota, and Breast Cancer: A Systematic Review. Life (Basel) 2024; 14:705. [PMID: 38929688 PMCID: PMC11204553 DOI: 10.3390/life14060705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/14/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Breast cancer is the most frequently diagnosed cancer in women worldwide. According to recent studies, alterations in the microbiota and epigenetic modulations are risk factors for this disease. This systematic review aims to determine the possible associations between the intestinal and mammary microbial populations, epigenetic modifications, and breast cancer. To achieve this objective, we conducted a literature search in the PubMed, Web of Science, and Science Direct databases following the PRISMA guidelines. Although no results are yet available in humans, studies in mice suggest a protective effect of maternal dietary interventions with bioactive compounds on the development of breast tumors in offspring. These dietary interventions also modified the gut microbiota, increasing the relative abundance of short-chain fatty acid-producing taxa and preventing mammary carcinogenesis. In addition, short-chain fatty acids produced by the microbiota act as epigenetic modulators. Furthermore, some authors indicate that stress alters the gut microbiota, promoting breast tumor growth through epigenetic and gene expression changes in the breast tumor microenvironment. Taken together, these findings show the ability of epigenetic modifications and alterations of the microbiota associated with environmental factors to modulate the development, aggressiveness, and progression of breast cancer.
Collapse
Affiliation(s)
- Alba Soldado-Gordillo
- Department of Biochemistry and Molecular Biology 2, School of Pharmacy, Campus de Cartuja s/n, 18071 Granada, Spain;
| | - Ana Isabel Álvarez-Mercado
- Department Pharmacology, School of Pharmacy, Campus de Cartuja s/n, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, Avda. del Conocimiento s/n, Armilla, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18071 Granada, Spain
| |
Collapse
|
5
|
Yin L, Zhang Q, Xie S, Cheng Z, Li R, Zhu H, Yu Q, Yuan H, Wang C, Peng H, Zhang G. HDAC inhibitor chidamide overcomes drug resistance in chronic myeloid leukemia with the T315i mutation through the Akt-autophagy pathway. Hum Cell 2023; 36:1564-1577. [PMID: 37222919 DOI: 10.1007/s13577-023-00919-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 05/16/2023] [Indexed: 05/25/2023]
Abstract
Currently, therapy for Chronic Myeloid Leukemia (CML) patients with the T315I mutation is a major challenge in clinical practice due to its high degree of resistance to first- and second-generation Tyrosine Kinase Inhibitors (TKIs). Chidamide, a Histone Deacetylase Inhibitor (HDACi) drug, is currently used to treat peripheral T-cell lymphoma. In this study, we investigated the anti-leukemia effects of chidamide on the CML cell lines Ba/F3 P210 and Ba/F3 T315I and primary tumor cells from CML patients with the T315I mutation. The underlying mechanism was investigated, and we found that chidamide could inhibit Ba/F3 T315I cells at G0/G1 phase. Signaling pathway analysis showed that chidamide induced H3 acetylation, downregulated pAKT expression and upregulated pSTAT5 expression in Ba/F3 T315I cells. Additionally, we found that the antitumor effect of chidamide could be exerted by regulating the crosstalk between apoptosis and autophagy. When chidamide was used in combination with imatinib or nilotinib, the antitumor effects were enhanced compared with chidamide alone in Ba/F3 T315I and Ba/F3 P210 cells. Therefore, we conclude that chidamide may overcome T315I mutation-related drug resistance in CML patients and works efficiently if used in combination with TKIs.
Collapse
MESH Headings
- Humans
- Histone Deacetylase Inhibitors/pharmacology
- Histone Deacetylase Inhibitors/therapeutic use
- Proto-Oncogene Proteins c-akt/genetics
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mutation
- Autophagy/genetics
- Apoptosis/genetics
- Cell Line, Tumor
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Cell Proliferation
Collapse
Affiliation(s)
- Le Yin
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Qingyang Zhang
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Sisi Xie
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Zhao Cheng
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Ruijuan Li
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Hongkai Zhu
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Qian Yu
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Huan Yuan
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| | - Canfei Wang
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China.
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China.
| | - Hongling Peng
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, 410011, Hunan, China.
- Institute of Molecular Hematology, Central South University, Changsha, China.
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China.
| | - Guangsen Zhang
- Division of Hematology, Second Xiang-Ya Hospital, Central South University, No.139th Renmin Middle Road, Changsha, 410011, Hunan, China
- Institute of Molecular Hematology, Central South University, Changsha, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, China
| |
Collapse
|
6
|
Keshawa Ediriweera M. Fatty acids as histone deacetylase inhibitors: old biochemistry tales in a new life sciences town. Drug Discov Today 2023; 28:103569. [PMID: 36990144 DOI: 10.1016/j.drudis.2023.103569] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Histone acetylation is a key epigenetic event. Although the keywords fatty acids, histones, and histone acetylation have a long history in biochemistry, these topics continue to attract much attention among researchers. The acetylation of histones is controlled by the activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs). An imbalance in the activities of HATs and HDACs is common in a range of human cancers. Histone deacetylase inhibitors (HDACi) can restore dysregulated histone acetylation profiles in cancer cells and have been identified as promising anti-cancer therapeutics. Short-chain fatty acids mediate anti-cancer effects by inhibiting the activity of HDACs. Recent studies have identified odd-chain fatty acids as novel HDACi. This review summarizes recent findings regarding fatty acids as HDACi in cancer therapy. Teaser: Inhibition of histone deacetylase (HDAC) activity by fatty acids.
Collapse
|
7
|
Giannattasio S, Dri M, Merra G, Caparello G, Rampello T, Di Renzo L. Effects of Fatty Acids on Hematological Neoplasms: A Mini Review. Nutr Cancer 2021; 74:1538-1548. [PMID: 34355630 DOI: 10.1080/01635581.2021.1960389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Hematological neoplasias are the fourth cause of death in the world. All of them are responsible of bad quality of life, due to heavy therapies administration and a lot of side effects correlated to. It arises a new concept of "multitherapy", in which fatty acids availment is used to contrast and reduce toxic effects and ameliorate chemotherapeutic agents asset. In Vitro studies have confirmed that fatty acids, in particular ω-3 eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are able to help canonical therapies to contrast cancer cell expansion and proliferation. In clinical trials it is also almost clear that fatty acids are useful to build new personalized therapies for a better condition of life. In this review we have summarized most recent studies on cancer cell lines and clinical trials on patients with fatty acids supplementation in diet therapies. We have found that fatty acids could be useful to contrast side effects during chemotherapeutic drugs therapies; they are also able to block cancer cell metabolic pathways for proliferation and contrast adverse effects, even when they are used in combination with traditional therapies or innovative, like monoclonal antibodies or CAR-T therapy. These aspects are crucial for better health condition of patients.
Collapse
Affiliation(s)
- Silvia Giannattasio
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Maria Dri
- Doctoral School of Applied Medical and Surgical Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Merra
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Giovanna Caparello
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Tiziana Rampello
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
8
|
Fattahi Y, Heidari HR, Khosroushahi AY. Review of short-chain fatty acids effects on the immune system and cancer. FOOD BIOSCI 2020. [DOI: 10.1016/j.fbio.2020.100793] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
9
|
Chen M, Jiang W, Xiao C, Yang W, Qin Q, Mao A, Tan Q, Lian B, Wei C. Sodium Butyrate Combined with Docetaxel for the Treatment of Lung Adenocarcinoma A549 Cells by Targeting Gli1. Onco Targets Ther 2020; 13:8861-8875. [PMID: 32982280 PMCID: PMC7501530 DOI: 10.2147/ott.s252323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose This study is aimed to investigate the combined treating efficacy of sodium butyrate and docetaxel on proliferation and apoptosis of the lung adenocarcinoma A549 cell line based on Gli1 regulation in vitro and in vivo. Materials and Methods RNA interference method was used to overexpress Gli1 in A549 cells. Cells were treated with varying concentrations of sodium butyrate, docetaxel or both in combination. CCK-8, colony formation assay, Hoechst 33258 staining, flow cytometry and TUNEL assay were employed to detect proliferation, cell cycle and apoptosis. qRT-PCR and Western blot analysis were applied to detect the mRNA and protein expression of Gli1. In vivo tumorigenicity was detected by tumor transplantation in nude mice. Downstream protein levels of Gli1 were detected using Western blot assay. Results It was found that sodium butyrate or docetaxel alone, respectively, inhibited proliferation and promoted apoptosis of A549 cells in vitro and in vivo, while the combination of the two generated significantly higher responses, which were also effective in another lung adenocarcinoma cell line H1299. Furthermore, the combined therapy had an additive effect in suppressing Gli1 expression and regulating the expression of its downstream proteins that involve in proliferation, cell cycle and apoptosis of A549 cells in vitro and in vivo, including decreased protein expression of Ki-67, CDK1, CDK2, Cyclin D1, Bcl-2 and Survivin, and increased protein expression of Cyclin A, p21, Bax and cleaved-Caspase 3. On the other hand, Gli1 overexpression perceptibly reversed the above-mentioned additive effect in vitro and in vivo. Conclusion This study demonstrates that the combined therapy of sodium butyrate and docetaxel additively inhibits proliferation and promotes apoptosis of A549 lung adenocarcinoma cells via suppressing Gli1 expression in vitro and in vivo. Targeting Gli1 by the combined therapy may provide new insights into the therapeutic management of patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Maojian Chen
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Wei Jiang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Chanchan Xiao
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, People's Republic of China
| | - Weiping Yang
- Department of Ultrasound Diagnosis, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Qinghong Qin
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Anyun Mao
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Qixing Tan
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Bin Lian
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Changyuan Wei
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| |
Collapse
|
10
|
Zhang Y, Ye M, Huang F, Wang S, Wang H, Mou X, Wang Y. Oncolytic Adenovirus Expressing ST13 Increases Antitumor Effect of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand Against Pancreatic Ductal Adenocarcinoma. Hum Gene Ther 2020; 31:891-903. [PMID: 32475172 DOI: 10.1089/hum.2020.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Oncolytic adenoviruses (OAds) are promising agents for cancer therapy, representing a novel therapeutic strategy for pancreatic ductal adenocarcinoma (PDAC). However, there are challenges associated with the successful use of an OAd alone, involving the security of the viral vector and screening of an effective antitumor gene. In the present study, a novel OAd CD55-ST13-tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) was constructed in which the dual therapeutic genes ST13 and TRAIL were inserted, featuring the carcinoembryonic antigen (CEA) as a promoter to control E1A and deletion of the 55 kDa E1B gene. ST13, known as a colorectal cancer suppressor gene, exhibited lower expression in PDAC than in tumor-adjacent tissues and was associated with poor prognosis in PDAC patients. In vitro studies demonstrated that CD55-ST13-TRAIL was effective in promoting the expression of ST13 and TRAIL in CEA-positive pancreatic cancer cells. Moreover, CD55-ST13-TRAIL exhibited a synergistic effect toward tumor cell death compared with CD55-ST13 alone or CD55-TRAIL alone, and inhibited tumor cell proliferation and induced cell apoptosis dependent on caspase pathways in PDAC cells. Furthermore, xenograft experiments in a mouse model indicated that CD55-ST13-TRAIL significantly inhibited tumor growth and improved the survival of animals with xenografts. The findings demonstrate that oncolytic virotherapy under the control of the promoter CEA enables safe and efficient treatment of PDAC, and suggest that it represents a promising candidate for the treatment of metastatic diseases.
Collapse
Affiliation(s)
- Youni Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| | - Miaojuan Ye
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| | - Fang Huang
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, P.R. China
| | - Shibing Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, P.R. China
| | - Huiju Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, P.R. China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, P.R. China
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| |
Collapse
|
11
|
Comparative effect of sodium butyrate and sodium propionate on proliferation, cell cycle and apoptosis in human breast cancer cells MCF-7. Breast Cancer 2020; 27:696-705. [PMID: 32095987 DOI: 10.1007/s12282-020-01063-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/10/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Short-chain fatty acids (SCFAs) are ubiquitous lipids produced as a result of bacterial fermentation of dietary fiber. While their role in colorectal cancer is well known, the effect of SCFAs in breast cancer is poorly defined. OBJECTIVE To understand the various effects of SCFAs on breast carcinogenesis, we investigated the effect of sodium butyrate (NaB) and sodium propionate (NaP) in MCF-7 cell line. MATERIALS AND METHODS Cells were incubated with different concentrations of NaB or NaP for 24, 48, 72 or 96 h. Cell proliferation was assayed using MTT kit. Cell cycle was performed using propidium iodide staining then analyzed with a flow cytometer. Apoptosis was assessed by Hoechst technique and cell-cycle sub-G1 phase. RESULTS NaB and NaP inhibited MCF-7 cell proliferation in a dose-dependent manner with respective IC50 of 1.26 mM and 4.5 mM, thus indicating that NaB is more potent than NaP. Low and medium levels of both SCFAs induced morphology changes which are characteristic of a differentiated phenotype. Flow cytometry analysis revealed a blockage in G1 growth phase. Interestingly, removing NaB or NaP from culture media after few days of treatment showed a reversible effect on cell morphology and proliferation where cells reentered the cycle after 24 h of drug wash-out. Finally, treatment with medium levels of these molecules induced low MCF-7 apoptosis, while higher doses led to massive apoptosis. CONCLUSION Our results show that SCFAs may be considered as an interesting inhibitor for breast cancer progression.
Collapse
|
12
|
Epigenetic mechanisms underlying the therapeutic effects of HDAC inhibitors in chronic myeloid leukemia. Biochem Pharmacol 2019; 173:113698. [PMID: 31706847 DOI: 10.1016/j.bcp.2019.113698] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022]
Abstract
Chronic myeloid leukemia (CML) is a hematological disorder caused by the oncogenic BCR-ABL fusion protein in more than 90% of patients. Despite the striking improvements in the management of CML patients since the introduction of tyrosine kinase inhibitors (TKis), the appearance of TKi resistance and side effects lead to treatment failure, justifying the need of novel therapeutic approaches. Histone deacetylase inhibitors (HDACis), able to modulate gene expression patterns and important cellular signaling pathways through the regulation of the acetylation status of both histone and non-histone protein targets, have been reported to display promising anti-leukemic properties alone or in combination with TKis. This review summarizes pre-clinical and clinical studies that investigated the mechanisms underlying the anticancer potential of HDACis and discusses the rationale for a combination of HDACis with TKis as a therapeutic option in CML.
Collapse
|
13
|
Xin P, Xu W, Zhu X, Li C, Zheng Y, Zheng T, Cheng W, Peng Q. Protective autophagy or autophagic death: effects of BEZ235 on chronic myelogenous leukemia. Cancer Manag Res 2019; 11:7933-7951. [PMID: 31686909 PMCID: PMC6709803 DOI: 10.2147/cmar.s204472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/05/2019] [Indexed: 12/11/2022] Open
Abstract
Purpose To investigate the effects of BEZ235 on chronic myeloid leukemia (CML) cells. Methods MTS assay was used to detect the proliferation of CML cells. The proteins expression were detected by Western blot assay. The effects of BEZ235 on autophagy in CML cells were verified through transmission electron microscopy and evaluated by laser confocal microscopy. Annexin V-FITC/PI double staining flow cytometry was used to detect apoptosis. A xenograft model was established to observe the therapeutic effect of BEZ235 in vivo. Results BEZ235 could inhibit the proliferation of CML cells; CQ and 3-MA could increase the proliferation inhibition and Z-VAD-FMK can reduce the proliferation inhibition of BEZ235 on CML cells (P<0.05). Results of TEM showed that the autophagosomes of CML cells treated with BEZ235 increased (P<0.05). The results by confocal microscopy showed that the autophagic activity of K562 cells increased with BEZ235 treatment. When BEZ235 combined with CQ, BEZ235-induced autophagic flow was blocked. FCM results showed that BEZ235 could induces apoptosis in CML cells. Z-VAD-FMK could decrease the apoptosis of CML cells induced by BEZ235. CQ increased the apoptosis of CML cells induced by BEZ235 (P<0.05). Western blot showed that BEZ235 inhibited the phosphorylation of AKT and S6K. BEZ235 alone could upregulate the expression of cleaved caspase-3 and LC3II. When combined with Z-VAD-FMK, the expression of cleaved caspase-3 was lower than that of BEZ235 alone. When combined with CQ, the expression of cleaved caspase-3 and LC3II were higher than those of BEZ235 alone (P<0.05). BEZ235 could inhibit the growth of xenografts of CML cell line. Conclusion BEZ235 can inhibit the proliferation of CML cells, induce apoptosis, and enhance autophagy activity. It induces protective autophagy. The combination of CQ can enhance the apoptosis and proliferation inhibition of CML cells induced by BEZ235.
Collapse
Affiliation(s)
- Pengliang Xin
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Wenqian Xu
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Xiongpeng Zhu
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Chuntuan Li
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Yan Zheng
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Tingjin Zheng
- Central Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Wenzhao Cheng
- Stem Cell Translational Research Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, People's Republic of China
| | - Qunyi Peng
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| |
Collapse
|
14
|
Jia X, Zheng Y, Guo Y, Chen K. Sodium butyrate and panobinostat induce apoptosis of chronic myeloid leukemia cells via multiple pathways. Mol Genet Genomic Med 2019; 7:e613. [PMID: 30891950 PMCID: PMC6503025 DOI: 10.1002/mgg3.613] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 01/05/2019] [Accepted: 02/10/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose Histone deacetylase inhibitor (HDACI) is a novel therapeutic option for cancer. However, the effects of HDACIs on chronic myeloid leukemia (CML) and the underlying mechanisms are still unknown. The aim of this study was to investigate the effect and the mechanism‐of‐action of two HDACI members, sodium butyrate (NaBu) and panobinostat (LBH589) in K562 and the adriamycin–resistant cell line K562/ADR. Methods Cell viability was assessed using MTT assay. Cell apoptosis was detected with flow cytometry. Cell cycle analysis and western blot were performed to explore the possible molecules related to HDACIs effects. Results The effect of NaBu was more powerful on K562/ADR than on K562 cells. LBH589 triggered apoptosis and inhibited the growth of K562 cells. Both HDACIs inhibited K562 and K562/ADR cells via activation of intrinsic/extrinsic apoptotic pathways and inhibition of AKT‐mTOR pathway while NaBu also activated endoplasmic reticulum stress (ERS) mediated apoptotic pathway in K562/ADR cells. LBH589 reduced the expression of drug–resistant related proteins in K562 cells. However, neither NaBu nor LBH589 could significantly influence the expression of the drug–resistant related proteins in K562/ADR cells. Conclusion The combination of HDACI and other therapeutic strategies are likely required to overcome drug resistance in CML therapy.
Collapse
Affiliation(s)
- Xiaoyuan Jia
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yinsuo Zheng
- Department of Hematology, Baoji Central Hospital, Baoji, China
| | - Yanzi Guo
- The Second Affiliated Hospital of Shaanxi Traditional University, Xianyang, China
| | - Kan Chen
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|