51
|
Li MX, Wu XT, Jing WQ, Hou WK, Hu S, Yan W. Inosine enhances tumor mitochondrial respiration by inducing Rag GTPases and nascent protein synthesis under nutrient starvation. Cell Death Dis 2023; 14:492. [PMID: 37532694 PMCID: PMC10397262 DOI: 10.1038/s41419-023-06017-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
Metabolic heterogeneity of tumor microenvironment (TME) is a hallmark of cancer and a big barrier to cancer treatment. Cancer cells display diverse capacities to utilize alternative carbon sources, including nucleotides, under poor nutrient circumstances. However, whether and how purine, especially inosine, regulates mitochondrial metabolism to buffer nutrient starvation has not been well-defined yet. Here, we identify the induction of 5'-nucleotidase, cytosolic II (NT5C2) gene expression promotes inosine accumulation and maintains cancer cell survival in the nutrient-poor region. Inosine elevation further induces Rag GTPases abundance and mTORC1 signaling pathway by enhancing transcription factor SP1 level in the starved tumor. Besides, inosine supplementary stimulates the synthesis of nascent TCA cycle enzymes, including citrate synthesis (CS) and aconitase 1 (ACO1), to further enhance oxidative phosphorylation of breast cancer cells under glucose starvation, leading to the accumulation of iso-citric acid. Inhibition of the CS activity or knockdown of ACO1 blocks the rescue effect of inosine on cancer survival under starvation. Collectively, our finding highlights the vital signal role of inosine linking mitochondrial respiration and buffering starvation, beyond serving as direct energy carriers or building blocks for genetic code in TME, shedding light on future cancer treatment by targeting inosine metabolism.
Collapse
Affiliation(s)
- Mei-Xin Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xiao-Ting Wu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Wen-Qiang Jing
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Wen-Kui Hou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Sheng Hu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Wei Yan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China.
| |
Collapse
|
52
|
Mirzaei S, Ranjbar B, Tackallou SH, Aref AR. Hypoxia inducible factor-1α (HIF-1α) in breast cancer: The crosstalk with oncogenic and onco-suppressor factors in regulation of cancer hallmarks. Pathol Res Pract 2023; 248:154676. [PMID: 37454494 DOI: 10.1016/j.prp.2023.154676] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Low oxygen level at tumor microenvironment leads to a condition, known as hypoxia that is implicated in cancer progression. Upon hypoxia, HIF-1α undergoes activation and due to its oncogenic function and interaction with other molecular pathways, promotes tumor progression. The HIF-1α role in regulating breast cancer progression is described, Overall, HIF-1α has upregulation in breast tumor and due to its tumor-promoting function, its upregulation is in favor of breast tumor progression. HIF-1α overexpression prevents apoptosis in breast tumor and it promotes cell cycle progression. Silencing HIF-1α triggers cycle arrest and decreases growth. Migration of breast tumor enhances by HIF-1α signaling and it mainly induces EMT in providing metastasis. HIF-1α upregulation stimulates drug resistance and radio-resistance in breast tumor. Furthermore, HIF-1α signaling induces immune evasion of breast cancer. Berberine and pharmacological intervention suppress HIF-1α signaling in breast tumor and regulation of HIF-1α by non-coding RNAs occurs. Furthermore, HIF-1α is a biomarker in clinic.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Bijan Ranjbar
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14117-13116, Iran
| | | | - Amir Reza Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
53
|
Kao TW, Bai GH, Wang TL, Shih IM, Chuang CM, Lo CL, Tsai MC, Chiu LY, Lin CC, Shen YA. Novel cancer treatment paradigm targeting hypoxia-induced factor in conjunction with current therapies to overcome resistance. J Exp Clin Cancer Res 2023; 42:171. [PMID: 37460927 DOI: 10.1186/s13046-023-02724-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/29/2023] [Indexed: 07/20/2023] Open
Abstract
Chemotherapy, radiotherapy, targeted therapy, and immunotherapy are established cancer treatment modalities that are widely used due to their demonstrated efficacy against tumors and favorable safety profiles or tolerability. Nevertheless, treatment resistance continues to be one of the most pressing unsolved conundrums in cancer treatment. Hypoxia-inducible factors (HIFs) are a family of transcription factors that regulate cellular responses to hypoxia by activating genes involved in various adaptations, including erythropoiesis, glucose metabolism, angiogenesis, cell proliferation, and apoptosis. Despite this critical function, overexpression of HIFs has been observed in numerous cancers, leading to resistance to therapy and disease progression. In recent years, much effort has been poured into developing innovative cancer treatments that target the HIF pathway. Combining HIF inhibitors with current cancer therapies to increase anti-tumor activity and diminish treatment resistance is one strategy for combating therapeutic resistance. This review focuses on how HIF inhibitors could be applied in conjunction with current cancer treatments, including those now being evaluated in clinical trials, to usher in a new era of cancer therapy.
Collapse
Affiliation(s)
- Ting-Wan Kao
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Geng-Hao Bai
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei City, 100225, Taiwan
| | - Tian-Li Wang
- Departments of Pathology, Oncology and Gynecology and Obstetrics, Johns Hopkins Medical Institutions, 1550 Orleans StreetRoom 306, Baltimore, MD, CRB221231, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ie-Ming Shih
- Departments of Pathology, Oncology and Gynecology and Obstetrics, Johns Hopkins Medical Institutions, 1550 Orleans StreetRoom 306, Baltimore, MD, CRB221231, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chi-Mu Chuang
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, 112304, Taiwan
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
- Department of Midwifery and Women Health Care, National Taipei University of Nursing and Health Sciences, Taipei, 112303, Taiwan
| | - Chun-Liang Lo
- Department of Biomedical Engineering, National Yang-Ming Chiao Tung University, Taipei, 112304, Taiwan
- Medical Device Innovation and Translation Center, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Meng-Chen Tsai
- Department of General Medicine, Taipei Medical University Hospital, Taipei, 110301, Taiwan
| | - Li-Yun Chiu
- Department of General Medicine, Mackay Memorial Hospital, Taipei, 104217, Taiwan
| | - Chu-Chien Lin
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- School of Medicine, College of Medicine, Taipei Medical University, Taipei City, 110301, Taiwan
| | - Yao-An Shen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
| |
Collapse
|
54
|
Srivastava N, Usmani SS, Subbarayan R, Saini R, Pandey PK. Hypoxia: syndicating triple negative breast cancer against various therapeutic regimens. Front Oncol 2023; 13:1199105. [PMID: 37492478 PMCID: PMC10363988 DOI: 10.3389/fonc.2023.1199105] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/05/2023] [Indexed: 07/27/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the deadliest subtypes of breast cancer (BC) for its high aggressiveness, heterogeneity, and hypoxic nature. Based on biological and clinical observations the TNBC related mortality is very high worldwide. Emerging studies have clearly demonstrated that hypoxia regulates the critical metabolic, developmental, and survival pathways in TNBC, which include glycolysis and angiogenesis. Alterations to these pathways accelerate the cancer stem cells (CSCs) enrichment and immune escape, which further lead to tumor invasion, migration, and metastasis. Beside this, hypoxia also manipulates the epigenetic plasticity and DNA damage response (DDR) to syndicate TNBC survival and its progression. Hypoxia fundamentally creates the low oxygen condition responsible for the alteration in Hypoxia-Inducible Factor-1alpha (HIF-1α) signaling within the tumor microenvironment, allowing tumors to survive and making them resistant to various therapies. Therefore, there is an urgent need for society to establish target-based therapies that overcome the resistance and limitations of the current treatment plan for TNBC. In this review article, we have thoroughly discussed the plausible significance of HIF-1α as a target in various therapeutic regimens such as chemotherapy, radiotherapy, immunotherapy, anti-angiogenic therapy, adjuvant therapy photodynamic therapy, adoptive cell therapy, combination therapies, antibody drug conjugates and cancer vaccines. Further, we also reviewed here the intrinsic mechanism and existing issues in targeting HIF-1α while improvising the current therapeutic strategies. This review highlights and discusses the future perspectives and the major alternatives to overcome TNBC resistance by targeting hypoxia-induced signaling.
Collapse
Affiliation(s)
- Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Salman Sadullah Usmani
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Rajasekaran Subbarayan
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
- Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Educations, Chennai, India
| | - Rashmi Saini
- Department of Zoology, Gargi College, University of Delhi, New Delhi, India
| | - Pranav Kumar Pandey
- Dr. R.P. Centre for Opthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
55
|
Michalkova R, Mirossay L, Kello M, Mojzisova G, Baloghova J, Podracka A, Mojzis J. Anticancer Potential of Natural Chalcones: In Vitro and In Vivo Evidence. Int J Mol Sci 2023; 24:10354. [PMID: 37373500 DOI: 10.3390/ijms241210354] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/12/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
There is no doubt that significant progress has been made in tumor therapy in the past decades. However, the discovery of new molecules with potential antitumor properties still remains one of the most significant challenges in the field of anticancer therapy. Nature, especially plants, is a rich source of phytochemicals with pleiotropic biological activities. Among a plethora of phytochemicals, chalcones, the bioprecursors of flavonoid and isoflavonoids synthesis in higher plants, have attracted attention due to the broad spectrum of biological activities with potential clinical applications. Regarding the antiproliferative and anticancer effects of chalcones, multiple mechanisms of action including cell cycle arrest, induction of different forms of cell death and modulation of various signaling pathways have been documented. This review summarizes current knowledge related to mechanisms of antiproliferative and anticancer effects of natural chalcones in different types of malignancies including breast cancers, cancers of the gastrointestinal tract, lung cancers, renal and bladder cancers, and melanoma.
Collapse
Affiliation(s)
- Radka Michalkova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Gabriela Mojzisova
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Janette Baloghova
- Department of Dermatovenerology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Anna Podracka
- Department of Dermatovenerology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| |
Collapse
|
56
|
Zhu Y, Wang S, Niu P, Chen H, Zhou J, Jiang L, Li D, Shi D. Raptor couples mTORC1 and ERK1/2 inhibition by cardamonin with oxidative stress induction in ovarian cancer cells. PeerJ 2023; 11:e15498. [PMID: 37304865 PMCID: PMC10257395 DOI: 10.7717/peerj.15498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Background A balance on nutrient supply and redox homeostasis is required for cell survival, and increased antioxidant capacity of cancer cells may lead to chemotherapy failure. Objective To investigate the mechanism of anti-proliferation of cardamonin by inducing oxidative stress in ovarian cancer cells. Methods After 24 h of drug treatment, CCK8 kit and wound healing test were used to detect cell viability and migration ability, respectively, and the ROS levels were detected by flow cytometry. The differential protein expression after cardamonin administration was analyzed by proteomics, and the protein level was detected by Western blotting. Results Cardamonin inhibited the cell growth, which was related to ROS accumulation. Proteomic analysis suggested that MAPK pathway might be involved in cardamonin-induced oxidative stress. Western blotting showed that cardamonin decreased Raptor expression and the activity of mTORC1 and ERK1/2. Same results were observed in Raptor KO cells. Notably, in Raptor KO cells, the effect of cardamonin was weakened. Conclusion Raptor mediated the function of cardamonin on cellular redox homeostasis and cell proliferation through mTORC1 and ERK1/2 pathways.
Collapse
|
57
|
Wenzel CK, von Montfort C, Ebbert L, Klahm NP, Reichert AS, Stahl W, Brenneisen P. The natural chalcone cardamonin selectively induces apoptosis in human neuroblastoma cells. Toxicol In Vitro 2023:105625. [PMID: 37268255 DOI: 10.1016/j.tiv.2023.105625] [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: 03/09/2023] [Revised: 05/11/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023]
Abstract
Neuroblastoma is the most common extracranial malignant tumor in childhood. Approximately 60% of all patients are classified as high-risk and require intensive treatment including non-selective chemotherapeutic agents leading to severe side effects. Recently, phytochemicals like the natural chalcone cardamonin (CD) have gained attention in cancer research. For the first time, we investigated the selective anti-cancer effects of CD in SH-SY5Y human neuroblastoma cells compared to healthy (normal) fibroblasts (NHDF). Our study revealed selective and dose-dependent cytotoxicity of CD in SH-SY5Y. The natural chalcone CD specifically altered the mitochondrial membrane potential (ΔΨm), as an early marker of apoptosis, in human neuroblastoma cells. Caspase activity was also selectively induced and the amount of cleaved caspase substrates such as PARP was thus increased in human neuroblastoma cells. CD-mediated apoptotic cell death was rescued by pan caspase inhibitor Z-VAD-FMK. The natural chalcone CD selectively induced apoptosis, the programmed cell death, in SH-SY5Y human neuroblastoma cells whereas NHDF being a model for normal (healthy) cells were unaffected. Our data indicates a clinical potential of CD in the more selective and less harmful treatment of neuroblastoma.
Collapse
Affiliation(s)
- Chantal-Kristin Wenzel
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Claudia von Montfort
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lara Ebbert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Niklas P Klahm
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Wilhelm Stahl
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Peter Brenneisen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
58
|
Yu YX, Wang S, Liu ZN, Zhang X, Hu ZX, Dong HJ, Lu XY, Zheng JB, Cui HJ. Traditional Chinese medicine in the era of immune checkpoint inhibitor: theory, development, and future directions. Chin Med 2023; 18:59. [PMID: 37210537 DOI: 10.1186/s13020-023-00751-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/13/2023] [Indexed: 05/22/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer management and have been widely applied; however, they still have some limitations in terms of efficacy and toxicity. There are multiple treatment regimens in Traditional Chinese Medicine (TCM) that play active roles in combination with Western medicine in the field of oncology treatment. TCM with ICIs works by regulating the tumor microenvironment and modulating gut microbiota. Through multiple targets and multiple means, TCM enhances the efficacy of ICIs, reverses resistance, and effectively prevents and treats ICI-related adverse events based on basic and clinical studies. However, there have been few conclusions on this topic. This review summarizes the development of TCM in cancer treatment, the mechanisms underlying the combination of TCM and ICIs, existing studies, ongoing trials, and prospects for future development.
Collapse
Affiliation(s)
- Yi-Xuan Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Shuo Wang
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Zhe-Ning Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Xu Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Zi-Xin Hu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Hui-Jing Dong
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Xing-Yu Lu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Jia-Bin Zheng
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China.
| | - Hui-Juan Cui
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, No.2 Yinghua East Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
59
|
Fan P, Meng H, Hao W, Zheng Y, Li H, Zhang Z, Du L, Guo X, Wang D, Wang Y, Wu H. Cardamonin targets KEAP1/NRF2 signaling for protection against atherosclerosis. Food Funct 2023; 14:4905-4920. [PMID: 37157847 DOI: 10.1039/d3fo00967j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Atherosclerosis (AS)-induced cardiovascular disease is a leading cause of death worldwide. To date, there is still a lack of effective approaches for AS intervention. Cardamonin (CAD) is a bioactive food component, but its effect on AS is unknown. In this work, CAD was investigated for its effect on AS using low-density lipoprotein receptor knockout mice and tumor necrosis factor-alpha (TNF-α)-stimulated endothelial cells (ECs). After a 12-week intervention, CAD was found to significantly prevent AS formation in the aortic root and aortic tree, reduce the necrotic core area, and inhibit aortic inflammation and oxidative stress. Moreover, CAD quenched TNF-α-provoked inflammation and oxidative stress in ECs. RNA-sequencing identified nuclear factor erythroid-2 related factor 2 (NFE2L2, NRF2)/heme oxidase 1 (HO1) signaling to be drastically activated by CAD. CAD is a known activator of the aryl hydrocarbon receptor (AHR) which is a transcription factor of the NFE2L2 gene. Surprisingly, AHR was not required for CAD's action on the activation of NRF2/HO1 signaling since AHR gene silencing did not reverse this effect. Furthermore, a molecular docking assay showed a strong binding potential of CAD to the Kelch domain of the Kelch-like ECH-associated protein 1 (KEAP1) which sequesters NRF2 in the cytoplasm. Both CAD and the Kelch domain inhibitor Ki696 promoted NRF2 nuclear translocation, whereas the combination of CAD and Ki696 did not yield a greater effect compared with either CAD or Ki696, confirming the interaction of CAD with the Kelch domain. This work provides an experimental basis for CAD as a novel and effective bioactive food component in future AS interventions.
Collapse
Affiliation(s)
- Pengfei Fan
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Huali Meng
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Wenhao Hao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Yan Zheng
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Hui Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Lei Du
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Xin Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Dongliang Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), 74 Zhongshan Road II, Guangzhou 510080, China
| | - Yunyan Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Rd., Jinan, Shandong 250012, China.
| | - Hao Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| |
Collapse
|
60
|
Liu B, Qu X, Wang J, Xu L, Zhang L, Xu B, Su J, Bian X. LINC00365 functions as a tumor suppressor by inhibiting HIF-1α-mediated glucose metabolism reprogramming in breast cancer. Exp Cell Res 2023; 425:113514. [PMID: 36804531 DOI: 10.1016/j.yexcr.2023.113514] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Long non-coding RNAs (lncRNAs) play an important role in regulating several physiological processes and have been implicated in several pathologies including cancer. LncRNAs have been found to regulate key cellular pathways involved in cancer development, and their aberrant expression plays critical roles in the onset or progression of disease. The role of lncRNAs in breast cancer (BC) has become a hot topic of research in recent years. We previously showed that LINC00365 inhibits BC survival. In the current study, based on the important role of energy metabolism and HIF-1α for tumor cell proliferation, we investigated the role and mechanism of the LINC00365/HIF-1α axis in affecting tumor growth through glycolysis using the breast cancer cell lines MCF-7 and HCC-1937. We found that LINC00365 inhibited BC cell proliferation. Furthermore, LINC00365 overexpression suppressed aerobic glycolysis in BC cells. RNA-sequencing identified hypoxia-inducible factor-1α (HIF-1α), which has been linked with glycolysis and upregulates glycolysis-related genes, as a potential target gene of LINC00365. Accordingly, we found that LINC00365 overexpression resulted in decreased expression of key glycolytic enzymes such as downstream hexokinase 2 (HK2), recombinant pyruvate kinase isozymes M2 (PKM2) and lactate dehydrogenase A (LDHA). Our results suggest that targeting LINC00365 may reverse the glucose metabolism pattern of BC and effectively inhibit BC survival both in vitro and in vivo.
Collapse
Affiliation(s)
- Buhan Liu
- Department of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Jilin Provincial Key Labor-atory of Surgical Translational Medicine, Changchun, China; Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xianzhi Qu
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, 130041, China
| | - Jian Wang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Long Xu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Lichao Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Bo Xu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jing Su
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xuehai Bian
- Department of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Jilin Provincial Key Labor-atory of Surgical Translational Medicine, Changchun, China.
| |
Collapse
|
61
|
Yang HL, Lin PY, Vadivalagan C, Lin YA, Lin KY, Hseu YC. Coenzyme Q 0 defeats NLRP3-mediated inflammation, EMT/metastasis, and Warburg effects by inhibiting HIF-1α expression in human triple-negative breast cancer cells. Arch Toxicol 2023; 97:1047-1068. [PMID: 36847822 DOI: 10.1007/s00204-023-03456-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/02/2023] [Indexed: 03/01/2023]
Abstract
Coenzyme Q0 (CoQ0) is a derivative quinone from Antrodia camphorata (AC) that exerts anticancer activities. This study examined the anticancer attributes of CoQ0 (0-4 µM) on inhibited anti-EMT/metastasis and NLRP3 inflammasome, and altered Warburg effects via HIF-1α inhibition in triple-negative breast cancer (MDA-MB-231 and 468) cells. MTT assay, cell migration/invasion assays, Western blotting, immunofluorescence, metabolic reprogramming, and LC-ESI-MS were carried out to assess the therapy potential of CoQ0. CoQ0 inhibited HIF-1α expression and suppressed the NLRP3 inflammasome and ASC/caspase-1 expression, followed by downregulation of IL-1β and IL-18 expression in MDA-MB-231 and 468 cells. CoQ0 ameliorated cancer stem-like markers by decreasing CD44 and increasing CD24 expression. Notably, CoQ0 modulated EMT by upregulating the epithelial marker E-cadherin and downregulating the mesenchymal marker N-cadherin. CoQ0 inhibited glucose uptake and lactate accumulation. CoQ0 also inhibited HIF-1α downstream genes involved in glycolysis, such as HK-2, LDH-A, PDK-1, and PKM-2 enzymes. CoQ0 decreased extracellular acidification rate (ECAR), glycolysis, glycolytic capacity, and glycolytic reserve in MDA-MB-231 and 468 cells under normoxic and hypoxic (CoCl2) conditions. CoQ0 inhibited the glycolytic intermediates lactate, FBP, and 2/3-PG, and PEP levels. CoQ0 increased oxygen consumption rate (OCR), basal respiration, ATP production, maximal respiration, and spare capacity under normoxic and hypoxic (CoCl2) conditions. CoQ0 increased TCA cycle metabolites, such as citrate, isocitrate, and succinate. CoQ0 inhibited aerobic glycolysis and enhanced mitochondrial oxidative phosphorylation in TNBC cells. Under hypoxic conditions, CoQ0 also mitigated HIF-1α, GLUT1, glycolytic-related (HK-2, LDH-A, and PFK-1), and metastasis-related (E-cadherin, N-cadherin, and MMP-9) protein or mRNA expression in MDA-MB-231 and/or 468 cells. Under LPS/ATP stimulation, CoQ0 inhibited NLRP3 inflammasome/procaspase-1/IL-18 activation and NFκB/iNOS expression. CoQ0 also hindered LPS/ATP-stimulated tumor migration and downregulated LPS/ATP-stimulated N-cadherin and MMP-2/-9 expression. The present study revealed that suppression of HIF-1α expression caused by CoQ0 may contribute to inhibition of NLRP3-mediated inflammation, EMT/metastasis, and Warburg effects of triple-negative breast cancers.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition, College of Health Care, China Medical University, No. 100, Section 1, Jingmao Road, Beitun, Taichung, 406040, Taiwan
| | - Ping-Yu Lin
- Institute of Nutrition, College of Health Care, China Medical University, No. 100, Section 1, Jingmao Road, Beitun, Taichung, 406040, Taiwan
| | - Chithravel Vadivalagan
- Department of Cosmeceutics, College of Pharmacy, China Medical University, No. 100, Section 1, Jingmao Road, Beitun, Taichung, 406040, Taiwan
| | - Yi-An Lin
- Institute of Nutrition, College of Health Care, China Medical University, No. 100, Section 1, Jingmao Road, Beitun, Taichung, 406040, Taiwan
| | - Kai-Yuan Lin
- Department of Medical Research, Chi-Mei Medical Center, Tainan, 710, Taiwan
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan, 71710, Taiwan
| | - You-Cheng Hseu
- Department of Cosmeceutics, College of Pharmacy, China Medical University, No. 100, Section 1, Jingmao Road, Beitun, Taichung, 406040, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, 41354, Taiwan.
- Chinese Medicine Research Center, China Medical University, Taichung, 40402, Taiwan.
- Research Center of Chinese Herbal Medicine, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
62
|
Cardamonin alleviates chondrocytes inflammation and cartilage degradation of osteoarthritis by inhibiting ferroptosis via p53 pathway. Food Chem Toxicol 2023; 174:113644. [PMID: 36731815 DOI: 10.1016/j.fct.2023.113644] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023]
Abstract
Osteoarthritis (OA) is a common degenerative joint disease, mainly presented by the deterioration of articular cartilage. Amounts of data demonstrated this deterioration is composed of oxidative stress, pro-inflammation and chondrocyte death events. Ferroptosis is a novel form of cell death that differs from apoptosis and autophagy, recent studies have shown that chondrocyte ferroptosis contributes to the development of osteoarthritis. Cardamonin (CAD) has been demonstrated to possess antioxidant and anti-inflammatory properties in several diseases, whether CAD may influence the OA progression is still obscure. Therefore, we aimed to determine whether CAD alleviates chondrocyte ferroptosis and its effect on OA with potential mechanism. In this study, we found that inflammation, cartilage degradation and ferroptosis induced by interleukin-1β (IL-1β) were significantly alleviated by CAD. Moreover, the administration of the ferroptosis inhibitor, Deferoxamine (DFO) reversed the inflammatory and cartilage degradation effects of IL-1β as well. Chondrocyte mitochondrial morphology and function were alleviated by both CAD and DFO. We found that CAD increased collagen II, p53, SLC7A11 GPX4 expression and decreased MMP13, iNOS, COX2 expression in chondrocytes, further investigation showed that the P53 signaling pathway was involved. In vivo, intra-articular injection of CAD significantly ameliorated cartilage damage in a rat OA model, induced collagen II and SLC7A11 expression by immunohistochemistry. Our study proves that CAD ameliorated OA cartilage degradation by regulating ferroptosis via P53 signaling pathway, suggesting a potential role of CAD in OA treatment.
Collapse
|
63
|
Gao S, Zhang W, Ma J, Ni X. PHF6 recruits BPTF to promote HIF-dependent pathway and progression in YAP-high breast cancer. J Transl Med 2023; 21:220. [PMID: 36967443 PMCID: PMC10040131 DOI: 10.1186/s12967-023-04031-8] [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: 05/25/2022] [Accepted: 03/01/2023] [Indexed: 03/27/2023] Open
Abstract
Background Aberrant epigenetic remodeling events contribute to progression and metastasis of breast cancer (Bca). The specific mechanims that epigenetic factors rely on to mediate tumor aggressiveness remain unclear. We aimed to elucidate the roles of epigenetic protein PHF6 in breast tumorigenesis. Methods Published datasets and tissue samples with PHF6 staining were used to investigate the clinical relevance of PHF6 in Bca. CCK-8, clony formation assays were used to assess cell growth capacity. Cell migration and invasion abilities were measured by Transwell assay. The gene mRNA and protein levels were measured by quantitative real-time PCR and western blot. Chromatin immunoprecipitation (ChIP)-qPCR assays were used to investigate transcriptional relationships among genes. The Co-immunoprecipitation (Co-IP) assay was used to validate interactions between proteins. The CRISPR/Cas9 editing technology was used to construct double HIF knockout (HIF-DKO) cells. The subcutaneous xenograft model and orthotopic implantation tumor model were used to asess in vivo tumor growth. Results In this study, we utilized MTT assay to screen that PHF6 is required for Bca growth. PHF6 promotes Bca proliferation and migration. By analyzing The Cancer Genome Atlas breast cancer (TCGA-Bca) cohort, we found that PHF6 was significantly higher in tumor versus normal tissues. Mechanistically, PHF6 physically interacts with HIF-1α and HIF-2α to potentiate HIF-driven transcriptional events to initiate breast tumorigenesis. HIF-DKO abolished PHF6-mediated breast tumor growth, and PHF6 deficiency in turn impaired HIF transcriptional effects. Besides, hypoxia could also rely on YAP activation, but not HIF, to sustain PHF6 expressions in Bca cells. In addition, PHF6 recuits BPTF to mediate epigenetic remodeling to augment HIF transcriptional activity. Targeting PHF6 or BPTF inhibitor (AU1) is effective in mice models. Lastly, PHF6 correlated with HIF target gene expression in human breast tumors, which is an independent prognostic regulator. Conclusions Collectively, this study identified PHF6 as a prognostic epigenetic regulator for Bca, functioning as a HIF coactivator. The fundamental mechanisms underlying YAP/PHF6/HIF axis in breast tumors endowed novel epigenegtic targets for Bca treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-023-04031-8.
Collapse
Affiliation(s)
- Sheng Gao
- grid.459791.70000 0004 1757 7869Department of Breast, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004 China
| | - Wensheng Zhang
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, Key Laboratory of Reproduction Regulation of NPFPC and Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, 200438 China
| | - Jingjing Ma
- grid.412676.00000 0004 1799 0784Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210004 China
| | - Xiaojian Ni
- grid.413087.90000 0004 1755 3939Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032 China
- grid.413087.90000 0004 1755 3939Cancer Center, ZhongShan Hospital, Fudan University, Shanghai, 200032 China
| |
Collapse
|
64
|
Michalkova R, Kello M, Cizmarikova M, Bardelcikova A, Mirossay L, Mojzis J. Chalcones and Gastrointestinal Cancers: Experimental Evidence. Int J Mol Sci 2023; 24:ijms24065964. [PMID: 36983038 PMCID: PMC10059739 DOI: 10.3390/ijms24065964] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Colorectal (CRC) and gastric cancers (GC) are the most common digestive tract cancers with a high incidence rate worldwide. The current treatment including surgery, chemotherapy or radiotherapy has several limitations such as drug toxicity, cancer recurrence or drug resistance and thus it is a great challenge to discover an effective and safe therapy for CRC and GC. In the last decade, numerous phytochemicals and their synthetic analogs have attracted attention due to their anticancer effect and low organ toxicity. Chalcones, plant-derived polyphenols, received marked attention due to their biological activities as well as for relatively easy structural manipulation and synthesis of new chalcone derivatives. In this study, we discuss the mechanisms by which chalcones in both in vitro and in vivo conditions suppress cancer cell proliferation or cancer formation.
Collapse
Affiliation(s)
- Radka Michalkova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Martina Cizmarikova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Annamaria Bardelcikova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| |
Collapse
|
65
|
Zhang J, Chen J, Xu J, Xue C, Mao Z. Plant-derived compounds for treating autosomal dominant polycystic kidney disease. FRONTIERS IN NEPHROLOGY 2023; 3:1071441. [PMID: 37675342 PMCID: PMC10479581 DOI: 10.3389/fneph.2023.1071441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/12/2023] [Indexed: 09/08/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), the most common monogenic hereditary kidney disease, is the fourth leading cause of end-stage kidney disease worldwide. In recent years, significant progress has been made in delaying ADPKD progression with different kinds of chemical drugs, such as tolvaptan, rapamycin, and somatostatin. Meanwhile, numerous plant-derived compounds have been investigated for their beneficial effects on slowing ADPKD progression. Among them, saikosaponin-d, Ganoderma triterpenes, curcumin, ginkgolide B, steviol, resveratrol, Sparganum stoloniferum Buch.-Ham, Cordyceps sinensis, triptolide, quercitrin, naringin, cardamonin, gambogic acid, and olive leaf extract have been found to retard renal cyst development by inhibiting cell proliferation or promoting cell apoptosis in renal cyst-lining epithelial cells. Metformin, a synthesized compound derived from French lilac or goat's rue (Galega officinalis), has been proven to retard the progression of ADPKD. This review focuses on the roles and mechanisms of plant-derived compounds in treating ADPKD, which may constitute promising new therapeutics in the future.
Collapse
Affiliation(s)
- Jieting Zhang
- School of Medicine, Shanghai University, Shanghai, China
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jiaxin Chen
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jing Xu
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Cheng Xue
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhiguo Mao
- Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
66
|
Liu S, Li Y, Yuan M, Song Q, Liu M. Correlation between the Warburg effect and progression of triple-negative breast cancer. Front Oncol 2023; 12:1060495. [PMID: 36776368 PMCID: PMC9913723 DOI: 10.3389/fonc.2022.1060495] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/06/2022] [Indexed: 01/28/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is ineligible for hormonal therapy and Her-2-targeted therapy due to the negative expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor-2. Although targeted therapy and immunotherapy have been shown to attenuate the aggressiveness of TNBC partially, few patients have benefited from them. The conventional treatment for TNBC remains chemotherapy. Chemoresistance, however, impedes therapeutic progress over time, and chemotherapy toxicity increases the burden of cancer on patients. Therefore, introducing more advantageous TNBC treatment options is a necessity. Metabolic reprogramming centered on glucose metabolism is considered a hallmark of tumors. It is described as tumor cells tend to convert glucose to lactate even under normoxic conditions, a phenomenon known as the Warburg effect. Similar to Darwinian evolution, its emergence is attributed to the selective pressures formed by the hypoxic microenvironment of pre-malignant lesions. Of note, the Warburg effect does not disappear with changes in the microenvironment after the formation of malignant tumor phenotypes. Instead, it forms a constitutive expression mediated by mutations or epigenetic modifications, providing a robust selective survival advantage for primary and metastatic lesions. Expanding evidence has demonstrated that the Warburg effect mediates multiple invasive behaviors in TNBC, including proliferation, metastasis, recurrence, immune escape, and multidrug resistance. Moreover, the Warburg effect-targeted therapy has been testified to be feasible in inhibiting TNBC progression. However, not all TNBCs are sensitive to glycolysis inhibitors because TNBC cells flexibly switch their metabolic patterns to cope with different survival pressures, namely metabolic plasticity. Between the Warburg effect-targeted medicines and the actual curative effect, metabolic plasticity creates a divide that must be continuously researched and bridged.
Collapse
Affiliation(s)
| | | | | | - Qing Song
- *Correspondence: Min Liu, ; Qing Song,
| | - Min Liu
- *Correspondence: Min Liu, ; Qing Song,
| |
Collapse
|
67
|
Liu Z, Hayashi H, Matsumura K, Uemura N, Shiraishi Y, Sato H, Baba H. Biological and Clinical Impacts of Glucose Metabolism in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:cancers15020498. [PMID: 36672448 PMCID: PMC9856866 DOI: 10.3390/cancers15020498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/07/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer type as it is prone to metastases and is difficult to diagnose at an early stage. Despite advances in molecular detection, its clinical prognosis remains poor and it is expected to become the second leading cause of cancer-related deaths. Approximately 85% of patients develop glucose metabolism disorders, most commonly diabetes mellitus, within three years prior to their pancreatic cancer diagnosis. Diabetes, or glucose metabolism disorders related to PDAC, are typically associated with insulin resistance, and beta cell damage, among other factors. From the perspective of molecular regulatory mechanisms, glucose metabolism disorders are closely related to PDAC initiation and development and to late invasion and metastasis. In particular, abnormal glucose metabolism impacts the nutritional status and prognosis of patients with PDAC. Meanwhile, preliminary research has shown that metformin and statins are effective for the prevention or treatment of malignancies; however, no such effect has been shown in clinical trials. Hence, the causes underlying these conflicting results require further exploration. This review focuses on the clinical significance of glucose metabolism disorders in PDAC and the mechanisms behind this relationship, while also summarizing therapeutic approaches that target glycolysis.
Collapse
|
68
|
Identification and Validation of a Novel Glycolysis-Related Gene Signature for Predicting the Prognosis and Therapeutic Response in Triple-Negative Breast Cancer. Adv Ther 2023; 40:310-330. [PMID: 36316558 DOI: 10.1007/s12325-022-02330-y] [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: 07/19/2022] [Accepted: 09/21/2022] [Indexed: 01/21/2023]
Abstract
INTRODUCTION A high malignancy rate and poor prognosis are common problems with triple-negative breast cancer (TNBC). There is increasing evidence that glycolysis plays vital roles in tumorigenesis, tumor invasion, immune evasion, chemoresistance, and metastasis. However, a comprehensive analysis of the diagnostic and prognostic significance of glycolysis in TNBC is lacking. METHODS Transcriptomic and clinical data of TNBC patients were obtained from The Cancer Genome Atlas (TCGA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) databases, respectively. Glycolysis-related genes (GRGs) were collected from the Molecular Signatures Database (MSigDB). Differential comparative analysis was performed to obtain the differentially expressed (DE)-GRGs associated with TNBC. Based on the DE-GRGs, a glycolysis-related risk signature was established using Least Absolute Shrinkage and Selector Operation (LASSO) and multivariable Cox regression analyses. The prognostic value, tumor microenvironment, mutation status, and chemotherapy response of different risk groups were analyzed. An independent cohort from the METABRIC database was used for external validation. Furthermore, the expression patterns of five genes derived from the prognostic model were validated by quantitative real-time polymerase chain reaction (RT-qPCR). RESULTS The glycolysis-related prognostic signature included five genes (IFNG, ACSS2, IRS2, GFUS, and GAL3ST1) and predicted the prognosis of TNBC patients independent of clinical factors (p < 0.05). Patients were divided into high- and low-risk groups based on the median risk score. Compared to low-risk TNBC patients, high-risk patients had significantly decreased overall survival (HR = 2.718, p < 0.001). Receiver operating characteristic and calibration curves demonstrated that the model had high performance in terms of predicting survival and risk stratification. The results remained consistent after external verification. Additionally, the tumor immune microenvironment significantly differed between the risk groups. Low-risk TNBC patients had a better immunotherapy response than high-risk patients. High-risk TNBC patients with a poor prognosis may benefit from targeted therapy. CONCLUSIONS This study developed a novel glycolysis and prognosis-related (GRP) signature based on GRGs to predict the prognosis of TNBC patients, and may aid clinical decision-making for these patients.
Collapse
|
69
|
Liang Z, Li X, Chen X, Zhou J, Li Y, Peng J, Lin Z, Liu G, Zeng X, Li C, Hang L, Li H. Fe/MOF based platform for NIR laser induced efficient PDT/PTT of cancer. Front Bioeng Biotechnol 2023; 11:1156079. [PMID: 37064235 PMCID: PMC10098195 DOI: 10.3389/fbioe.2023.1156079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/15/2023] [Indexed: 04/18/2023] Open
Abstract
Introduction: Photodynamic therapy (PDT) and photothermal therapy (PTT) are widely used in the treatment of tumors. However, their application in the treatment of clinical tumors is limited by the complexity and irreversible hypoxia environment generated by tumor tissues. To overcome this limitation, a nanoparticle composed of indocyanine green (ICG) and Fe-MOF-5 was developed. Methods: We prepared F-I@FM5 and measured its morphology, particle size, and stability. Its enzyme like ability and optical effect was verified. Then we used MTT, staining and flow cytometry to evaluated the anti-tumor effect on EMT-6 cells in vitro. Finally, the anti-tumor effect in vivo has been studied on EMT-6 tumor bearing mice. Results: For the composite nanoparticle, we confirmed that Fe-MOF-5 has the best nanozyme activity. In addition, it has excellent photothermal conversion efficiency and generates reactive oxygen species (ROS) under near-infrared light irradiation (808 nm). The composite nanoparticle showed good tumor inhibition effect in vitro and in vivo, which was superior to the free ICG or Fe-MOF-5 alone. Besides, there was no obvious cytotoxicity in major organs within the effective therapeutic concentration. Discussion: Fe-MOF-5 has the function of simulating catalase, which can promote the decomposition of excessive H2O2 in the tumor microenvironment and produce oxygen to improve the hypoxic environment. The improvement of tumor hypoxia can enhance the efficacy of PDT and PTT. This research not only provides an efficient and stable anti-tumor nano platform, but also has broad application prospects in the field of tumor therapy, and provides a new idea for the application of MOF as an important carrier material in the field of photodynamic therapy.
Collapse
Affiliation(s)
- Zixing Liang
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaofeng Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaofang Chen
- Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiawei Zhou
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yanan Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Jianhui Peng
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zhousheng Lin
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Gai Liu
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiancheng Zeng
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Cheng Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
- Jinan University, Guangzhou, China
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Hainan, China
- *Correspondence: Hailiang Li, ; Cheng Li, ; Lifeng Hang,
| | - Lifeng Hang
- Guangdong Second Provincial General Hospital, Guangzhou, China
- *Correspondence: Hailiang Li, ; Cheng Li, ; Lifeng Hang,
| | - Hailiang Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
- *Correspondence: Hailiang Li, ; Cheng Li, ; Lifeng Hang,
| |
Collapse
|
70
|
Hu X, Guo B, Sun T, Wang W. Inhibition of glycolysis represses the growth and alleviates the endoplasmic reticulum stress of breast cancer cells by regulating TMTC3. Open Med (Wars) 2023; 18:20230635. [PMID: 37069941 PMCID: PMC10105522 DOI: 10.1515/med-2023-0635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/05/2022] [Accepted: 12/11/2022] [Indexed: 04/19/2023] Open
Abstract
Considering the role of glycolysis inhibition as a novel therapeutic strategy for cancer, including breast cancer (BC), we wondered whether glycolysis could affect BC progression by regulating transmembrane O-mannosyltransferase-targeting cadherins 3 (TMTC3). Following the intervention, lactic acid production in BC cells was monitored, and viability, proliferation, and apoptosis assays were performed. The expressions of TMTC3 and endoplasmic reticulum (ER) stress- and apoptosis-related factors Caspase-12, C/EBP homologous protein (CHOP), glucose-regulated protein 78 (GRP78), B-cell lymphoma-2 (Bcl-2), and Bcl-2 associated X (Bax) were quantified. TMTC3 was lowly expressed in BC tissue and cell. The promotion of glycolysis via glucose represses TMTC3 expression and apoptosis yet enhances lactic acid production and growth of BC cell, along with promoted levels of Caspase-12, CHOP, GRP78, and Bcl-2 yet repressed level of Bax, while the contrary results were evidenced after 2-deoxyglycouse intervention. Overexpressed TMTC3 additionally abrogated the effects of glycolysis on increasing the viability and proliferation yet inhibiting the apoptosis of BC cells, with the increased expressions of Caspase-12, CHOP, and GRP78, and Bcl-2 yet decreased level of Bax. Collectively, inhibiting glycolysis restrained the growth and attenuated the ER stress of BC cell by regulating TMTC3.
Collapse
Affiliation(s)
- Xue Hu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun City, Jilin Province, 130033, China
| | - Baoliang Guo
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, China
| | - Tong Sun
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun City, Jilin Province, 130033, China
| | - Wan Wang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, No. 126 Xiantai
Avenue, Nanguan District, Changchun City, Jilin Province, 130033, China
| |
Collapse
|
71
|
siRNA and targeted delivery systems in breast cancer therapy. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 25:1167-1188. [PMID: 36562927 DOI: 10.1007/s12094-022-03043-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
Recently, nucleic acid drugs have been considered as promising candidates in treatment of various diseases, especially cancer. Because of developing resistance to conventional chemotherapy, use of genetic tools in cancer therapy appears inevitable. siRNA is a RNAi tool with capacity of suppressing target gene. Owing to overexpression of oncogenic factors in cancer, siRNA can be used for suppressing those pathways. This review emphasizes the function of siRNA in treatment of breast tumor. The anti-apoptotic-related genes including Bcl-2, Bcl-xL and survivin can be down-regulated by siRNA in triggering cell death in breast cancer. STAT3, STAT8, Notch1, E2F3 and NF-κB are among the factors with overexpression in breast cancer that their silencing by siRNA paves the way for impairing tumor proliferation and invasion. The oncogenic mechanisms in drug resistance development in breast tumor such as lncRNAs can be suppressed by siRNA. Furthermore, siRNA reducing P-gp activity can increase drug internalization in tumor cells. Because of siRNA degradation at bloodstream and low accumulation at tumor site, nanoplatforms have been employed for siRNA delivery to suppress breast tumor progression via improving siRNA efficacy in gene silencing. Development of biocompatible and efficient nanostructures for siRNA delivery can make milestone progress in alleviation of breast cancer patients.
Collapse
|
72
|
Liu Q, Guan C, Liu C, Li H, Wu J, Sun C. Targeting hypoxia-inducible factor-1alpha: A new strategy for triple-negative breast cancer therapy. Biomed Pharmacother 2022; 156:113861. [DOI: 10.1016/j.biopha.2022.113861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/02/2022] Open
|
73
|
Zheng X, Ma H, Wang J, Huang M, Fu D, Qin L, Yin Q. Energy metabolism pathways in breast cancer progression: The reprogramming, crosstalk, and potential therapeutic targets. Transl Oncol 2022; 26:101534. [PMID: 36113343 PMCID: PMC9482139 DOI: 10.1016/j.tranon.2022.101534] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/14/2022] [Accepted: 09/04/2022] [Indexed: 11/19/2022] Open
Abstract
Breast cancer (BC) is a malignant tumor that seriously endangers health in women. BC, like other cancers, is accompanied by metabolic reprogramming. Among energy metabolism-related pathways, BC exhibits enhanced glycolysis, tricarboxylic acid (TCA) cycle, pentose phosphate pathway (PPP), glutamate metabolism, and fatty acid metabolism activities. These pathways facilitate the proliferation, growth and migration of BC cells. The progression of BC is closely related to the alterations in the activity or expression level of several metabolic enzymes, which are regulated by the intrinsic factors such as the key signaling and transcription factors. The metabolic reprogramming in the progression of BC is attributed to the aberrant expression of the signaling and transcription factors associated with the energy metabolism pathways. Understanding the metabolic mechanisms underlying the development of BC will provide a druggable potential for BC treatment and drug discovery.
Collapse
Affiliation(s)
- Xuewei Zheng
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Haodi Ma
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jingjing Wang
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Mengjiao Huang
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Dongliao Fu
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Ling Qin
- Department of Hematology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Qinan Yin
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
74
|
Lu T, Zheng C, Fan Z. Cardamonin suppressed the migration, invasion, epithelial mesenchymal transition (EMT) and lung metastasis of colorectal cancer cells by down-regulating ADRB2 expression. PHARMACEUTICAL BIOLOGY 2022; 60:1011-1021. [PMID: 35645356 PMCID: PMC9154753 DOI: 10.1080/13880209.2022.2069823] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 03/09/2022] [Accepted: 04/11/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Cardamonin (CDN) can suppress cell growth in colorectal cancer (CRC), a common digestive malignancy. OBJECTIVE We explored the effect and mechanism of CDN on metastatic CRC. MATERIALS AND METHODS Two cell lines (HT29 and HCT116) were initially treated with CDN at different concentrations (5, 10 and 20 μmol/L) or 50 μmol/L propranolol (positive control) for 24 or 48 h. Then, the two cell lines were separately transfected with siADRB2 and ADRB2 overexpression plasmids, and further treated with 10 μmol/L CDN for 24 h. The cell viability, migration and invasion were determined by cell counting kit-8 (CCK-8), wound healing and transwell assays, respectively. The levels of ADRB2, matrix metalloprotease (MMP)-2, MMP-9, E-cadherin and N-cadherin were measured by Western blotting or/and RT-qPCR. A CRC metastasis model was established to evaluate the antimetastatic potential of CDN (25 mg/kg). RESULTS ADRB2 (3.2-fold change; p < 0.001) was highly expressed in CRC tissues. CDN at 10 μmol/L suppressed viability (69% and 70%), migration (33% and 66%), invasion (43% and 72%) and ADRB2 expression (2.2- and 2.84-fold change) in HT29 and HCT116 cells (p < 0.001). CDN at 10 μmol/L inhibited MMP-2, MMP-9 and N-cadherin expression but promoted E-cadherin expression in CRC cells (p < 0.001). Importantly, the effect of CDN on CRC cells was impaired by ADRB2 overexpression, but further enhanced by ADRB2 down-regulation (p < 0.01). Additionally, ADRB2 overexpression reversed the inhibitory effect of CDN on metastatic lung nodules (p < 0.05). Discussion and conclusions: CDN is a potential candidate for the treatment of metastatic CRC in clinical practice.
Collapse
Affiliation(s)
- Ting Lu
- Proctology Department, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunju Zheng
- Proctology Department, Huai’an TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Huai’an, China
| | - Zhimin Fan
- Proctology Department, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
75
|
Luo S, Jiang Y, Anfu Zheng, Zhao Y, Wu X, Li M, Du F, Chen Y, Deng S, Chen M, Li W, Li X, Gu L, Sun Y, Xiao Z, Shen J. Targeting hypoxia-inducible factors for breast cancer therapy: A narrative review. Front Pharmacol 2022; 13:1064661. [PMID: 36532768 PMCID: PMC9751339 DOI: 10.3389/fphar.2022.1064661] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/18/2022] [Indexed: 09/15/2023] Open
Abstract
Hypoxia-inducible factors (HIFs), central regulators for cells to adapt to low cellular oxygen levels, are often overexpressed and activated in breast cancer. HIFs modulate the primary transcriptional response of downstream pathways and target genes in response to hypoxia, including glycolysis, angiogenesis and metastasis. They can promote the development of breast cancer and are associated with poor prognosis of breast cancer patients by regulating cancer processes closely related to tumor invasion, metastasis and drug resistance. Thus, specific targeting of HIFs may improve the efficiency of cancer therapy. In this review, we summarize the advances in HIF-related molecular mechanisms and clinical and preclinical studies of drugs targeting HIFs in breast cancer. Given the rapid progression in this field and nanotechnology, drug delivery systems (DDSs) for HIF targeting are increasingly being developed. Therefore, we highlight the HIF related DDS, including liposomes, polymers, metal-based or carbon-based nanoparticles.
Collapse
Affiliation(s)
- Shuang Luo
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
- Department of Pharmacy, The Second People’s Hospital of Jiangyou, Mianyang, China
| | - Yu Jiang
- Department of Pharmacy, The People’s Hospital of Wusheng, Guang’an, China
| | - Anfu Zheng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Zhangang Xiao
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| |
Collapse
|
76
|
Wang Y, Xu Y, Guo W, Fang Y, Hu L, Wang R, Zhao R, Guo D, Qi B, Ren G, Ren J, Li Y, Zhang M. Ablation of Shank3 alleviates cardiac dysfunction in aging mice by promoting CaMKII activation and Parkin-mediated mitophagy. Redox Biol 2022; 58:102537. [PMID: 36436456 PMCID: PMC9709154 DOI: 10.1016/j.redox.2022.102537] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Compromised mitophagy and mitochondrial homeostasis are major contributors for the etiology of cardiac aging, although the precise underlying mechanisms remains elusive. Shank3, a heart-enriched protein, has recently been reported to regulate aging-related neurodegenerative diseases. This study aimed to examine the role of Shank3 in the pathogenesis of cardiac senescence and the possible mechanisms involved. Cardiac-specific conditional Shank3 knockout (Shank3CKO) mice were subjected to natural aging. Mitochondrial function and mitophagy activity were determined in vivo, in mouse hearts and in vitro, in cardiomyocytes. Here, we showed that cardiac Shank3 expression exhibited a gradual increase during the natural progression of the aging, accompanied by overtly decreased mitophagy activity and a decline in cardiac function. Ablation of Shank3 promoted mitophagy, reduced mitochondria-derived superoxide (H2O2 and O2•-) production and apoptosis, and protected against cardiac dysfunction in the aged heart. In an in vitro study, senescent cardiomyocytes treated with D-gal exhibited reduced mitophagy and significantly elevated Shank3 expression. Shank3 knock-down restored mitophagy, leading to increased mitochondrial membrane potential, decreased mitochondrial oxidative stress, and reduced apoptosis in senescent cardiomyocytes, whereas Shank3 overexpression mimicked D-gal-induced mitophagy inhibition and mitochondrial dysfunction in normally cultured cardiomyocytes. Mechanistically, the IP assay revealed that Shank3 directly binds to CaMKII, and this interaction was further increased in the aged heart. Enhanced Shank3/CaMKII binding impedes mitochondrial translocation of CaMKII, resulting in the inhibition of parkin-mediated mitophagy, which ultimately leads to mitochondrial dysfunction and cardiac damage in the aged heart. Our study identified Shank3 as a novel contributor to aging-related cardiac damage. Manipulating Shank3/CaMKII-induced mitophagy inhibition could thus be an optional strategy for therapeutic intervention in clinical aging-related cardiac dysfunctions.
Collapse
Affiliation(s)
- Ying Wang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Yuerong Xu
- Department of Orthodontics, School of Stomatology, Air Force Medical University, Xi’an, 710032, China
| | - Wangang Guo
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Yexian Fang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Lang Hu
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Runze Wang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Ran Zhao
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Dong Guo
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Bingchao Qi
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Gaotong Ren
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China,Corresponding author. Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China,Corresponding author.
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China,Corresponding author.
| |
Collapse
|
77
|
Yang Y, Wu H, Zou X, Chen Y, He R, Jin Y, Zhou B, Ge C, Yang Y. A novel synthetic chalcone derivative, 2,4,6-trimethoxy-4'-nitrochalcone (Ch-19), exerted anti-tumor effects through stimulating ROS accumulation and inducing apoptosis in esophageal cancer cells. Cell Stress Chaperones 2022; 27:645-657. [PMID: 36242757 PMCID: PMC9672279 DOI: 10.1007/s12192-022-01302-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 01/25/2023] Open
Abstract
Esophageal cancer has always been associated with poor prognosis and a low five-year survival rate. Chalcone, a flavonoid family member, has shown anti-tumor property in several types of cancer. However, few studies reported the potency and mechanisms of action of synthetic Chalcone derivatives against esophageal squamous cell carcinoma. In this study, we designed and synthesized a series of novel chalcone analogs and Ch-19 was selected for its superior anti-tumor potency. Results indicated that Ch-19 shows a dose- and time-dependent anti-tumor activity in both KYSE-450 and Eca-109 esophageal cancer cells. Moreover, treatment of Ch-19 resulted in the regression of KYSE-450 tumor xenografts in nude mice. Furthermore, we investigated the potential mechanism involved in the effective anti-tumor effects of Ch-19. As a result, we observed that Ch-19 treatment promoted ROS accumulation and caused G2/M phase arrest in both Eca-109 and KYSE-450 cancer cell lines, thereby resulting in cell apoptosis. Taken together, our study provided a novel synthetic chalcone derivative as a potential anti-tumor therapeutic candidate for treating esophageal cancer.
Collapse
Affiliation(s)
- Yan Yang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Department of Experimental Therapeutics, British Columbia Cancer Research Centre, University of British Columbia, Vancouver, Canada
| | - He Wu
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xiao Zou
- Department of Oncology and Hematology, The First People's Hospital of Taian, Taian, China
| | - Yongye Chen
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Runjia He
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yibo Jin
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Bei Zhou
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Chunpo Ge
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| | - Yun Yang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
78
|
Zhao Y, Chard Dunmall LS, Cheng Z, Wang Y, Si L. Natural products targeting glycolysis in cancer. Front Pharmacol 2022; 13:1036502. [PMID: 36386122 PMCID: PMC9663463 DOI: 10.3389/fphar.2022.1036502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/17/2022] [Indexed: 09/05/2023] Open
Abstract
Many energy metabolism pathways exist in cancer, including glycolysis, amino acid metabolism, fatty acid oxidation, and mitochondrial respiration. Tumor cells mainly generate energy through glycolysis to maintain growth and biosynthesis of tumor cells under aerobic conditions. Natural products regulate many steps in glycolysis and targeting glycolysis using natural products is a promising approach to cancer treatment. In this review, we exemplify the relationship between glycolysis and tumors, demonstrate the natural products that have been discovered to target glycolysis for cancer treatment and clarify the mechanisms involved in their actions. Natural products, such as resveratrol mostly found in red grape skin, licochalcone A derived from root of Glycyrrhiza inflate, and brusatol found in Brucea javanica and Brucea mollis, largely derived from plant or animal material, can affect glycolysis pathways in cancer by targeting glycolytic enzymes and related proteins, oncogenes, and numerous glycolytic signal proteins. Knowledge of how natural products regulate aerobic glycolysis will help illuminate the mechanisms by which these products can be used as therapeutics to inhibit cancer cell growth and regulate cellular metabolism. Systematic Review Registration: https://pubmed.ncbi.nlm.nih.gov/, https://clinicaltrials.gov/, http://lib.zzu.edu.cn/.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa S Chard Dunmall
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Zhenguo Cheng
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Lingling Si
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
79
|
Nrf2 Modulation in Breast Cancer. Biomedicines 2022; 10:biomedicines10102668. [PMID: 36289931 PMCID: PMC9599257 DOI: 10.3390/biomedicines10102668] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 12/05/2022] Open
Abstract
Reactive oxygen species (ROS) are identified to control the expression and activity of various essential signaling intermediates involved in cellular proliferation, apoptosis, and differentiation. Indeed, ROS represents a double-edged sword in supporting cell survival and death. Many common pathological processes, including various cancer types and neurodegenerative diseases, are inflammation and oxidative stress triggers, or even initiate them. Keap1-Nrf2 is a master antioxidant pathway in cytoprotective mechanisms through Nrf2 target gene expression. Activation of the Nfr2 pathway benefits cells in the early stages and reduces the level of ROS. In contrast, hyperactivation of Keap1-Nrf2 creates a context that supports the survival of both healthy and cancerous cells, defending them against oxidative stress, chemotherapeutic drugs, and radiotherapy. Considering the dual role of Nrf2 in suppressing or expanding cancer cells, determining its inhibitory/stimulatory position and targeting can represent an impressive role in cancer treatment. This review focused on Nrf2 modulators and their roles in sensitizing breast cancer cells to chemo/radiotherapy agents.
Collapse
|
80
|
Cui Y, Li C, Sang F, Cao W, Qin Z, Zhang P. Natural products targeting glycolytic signaling pathways-an updated review on anti-cancer therapy. Front Pharmacol 2022; 13:1035882. [PMID: 36339566 PMCID: PMC9631946 DOI: 10.3389/fphar.2022.1035882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/30/2022] [Indexed: 11/30/2022] Open
Abstract
Glycolysis is a complex metabolic process that occurs to convert glucose into pyruvate to produce energy for living cells. Normal cells oxidized pyruvate into adenosine triphosphate and carbon dioxide in the presence of oxygen in mitochondria while cancer cells preferentially metabolize pyruvate to lactate even in the presence of oxygen in order to maintain a slightly acidic micro-environment of PH 6.5 and 6.9, which is beneficial for cancer cell growth and metastasis. Therefore targeting glycolytic signaling pathways provided new strategy for anti-cancer therapy. Natural products are important sources for the treatment of diseases with a variety of pharmacologic activities. Accumulated studies suggested that natural products exhibited remarkable anti-cancer properties both in vitro and in vivo. Plenty of studies suggested natural products like flavonoids, terpenoids and quinones played anti-cancer properties via inhibiting glucose metabolism targets in glycolytic pathways. This study provided an updated overview of natural products controlling glycolytic pathways, which also provide insight into druggable mediators discovery targeting cancer glucose metabolism.
Collapse
Affiliation(s)
- Yuting Cui
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Chuang Li
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Feng Sang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Weiling Cao
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| | - Zhuo Qin
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| | - Peng Zhang
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| |
Collapse
|
81
|
Liu C, Li S, Zhang X, Jin C, Zhao B, Li L, Miao QR, Jin Y, Fan Z. Nogo-B receptor increases glycolysis and the paclitaxel resistance of estrogen receptor-positive breast cancer via the HIF-1α-dependent pathway. Cancer Gene Ther 2022; 30:647-658. [PMID: 36241702 DOI: 10.1038/s41417-022-00542-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 09/15/2022] [Accepted: 10/03/2022] [Indexed: 12/29/2022]
Abstract
Chemotherapy can improve the prognosis and overall survival of breast cancer patients, but chemoresistance continues a major problem in clinical. Most breast cancer is estrogen receptor (ER) positive but responds less to neoadjuvant or adjuvant chemotherapy than ER-negative breast cancer. The Nogo-B receptor (NgBR) increases the chemoresistance of ER-positive breast cancer by facilitating oncogene signaling pathways. Here, we further investigated the potential role of NgBR as a novel target to overcome glycolysis-dependent paclitaxel resistance in ER-positive breast cancer. NgBR knockdown inhibited glycolysis and promoted paclitaxel-induced apoptosis by attenuating HIF-1α expression in ER-positive breast cancer cells via NgBR-mediated estrogen receptor alpha (ERα)/hypoxia-inducible factor-1 alpha (HIF-1α) and nuclear factor-kappa B subunit (NF-κB)/HIF-1α signaling pathways. A ChIP assay further confirmed that NgBR overexpression not only facilitates ERα binding to HIF-1α and GLUT1 genes but also promotes HIF-1α binding to GLUT1, HK2, and LDHA genes, which further promotes glycolysis and induces paclitaxel resistance. In conclusion, our study suggests that NgBR expression is essential for maintaining the metabolism and paclitaxel resistance of ER-positive breast cancer, and the NgBR can be a new therapeutic target for improving chemoresistance in ER-positive breast cancer.
Collapse
Affiliation(s)
- Chang Liu
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Sijie Li
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoxiao Zhang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chunxiang Jin
- Institute Department of Ultrasonography, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Baofeng Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute, Liaoning, China
| | - Liying Li
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Qing Robert Miao
- Department of Foundations of Medicine, NYU Long Island School of Medicine, New York, NY, USA.
| | - Ying Jin
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Zhimin Fan
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
82
|
Correlation of Glucose Metabolism with Cancer and Intervention with Traditional Chinese Medicine. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2192654. [PMID: 36276846 PMCID: PMC9586738 DOI: 10.1155/2022/2192654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/17/2022] [Accepted: 09/10/2022] [Indexed: 11/07/2022]
Abstract
Cancer is a complex disease with several distinct characteristics, referred to as “cancer markers” one of which is metabolic reprogramming, which is a common feature that drives cancer progression. Over the last ten years, researchers have focused on the reprogramming of glucose metabolism in cancer. In cancer, the oxidative phosphorylation metabolic pathway is converted into the glycolytic pathway in order to meet the growth requirements of cancer cells, thereby creating a microenvironment that promotes cancer progression. The precise mechanism of glucose metabolism in cancer cells is still unknown, but it is thought to involve the aberrant levels of metabolic enzymes, the influence of the tumor microenvironment (TME), and the activation of tumor-promoting signaling pathways. It is suggested that glucose metabolism is strongly linked to cancer progression because it provides energy to cancer cells and interferes with antitumor drug pharmacodynamics. Therefore, it is critical to unravel the mechanism of glucose metabolism in tumors in order to gain a better understanding of tumorigenesis and to lay the groundwork for future research into the identification of novel diagnostic markers and therapeutic targets for cancer treatment. Traditional Chinese Medicine (TCM) has the characteristics of multiple targets, multiple components, and less toxic side effects and has unique advantages in tumor treatment. In recent years, researchers have found that a variety of Chinese medicine monomers and compound recipes play an antitumor role by interfering with the reprogramming of tumor metabolism. The underlying mechanisms of metabolism reprogramming of tumor cells and the role of TCM in regulating glucose metabolism are reviewed in this study, so as to provide a new idea for antitumor research in Chinese medicine.
Collapse
|
83
|
Chen H, Huang S, Niu P, Zhu Y, Zhou J, Jiang L, Li D, Shi D. Cardamonin suppresses pro-tumor function of macrophages by decreasing M2 polarization on ovarian cancer cells via mTOR inhibition. Mol Ther Oncolytics 2022; 26:175-188. [PMID: 35860007 PMCID: PMC9278033 DOI: 10.1016/j.omto.2022.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 06/22/2022] [Indexed: 12/28/2022] Open
Abstract
Ovarian cancer is the most fatal tumor characterized by an abundance of tumor-associated macrophage (TAM) infiltrations in women. Functional TAMs, which mainly present M2-like phenotypes and perform key functions on tumor progress, have been considered an attractive target for ovarian cancer therapy. Cardamonin showed an excellent antitumor activity in multiple tumor cells. This study aimed to investigate the role of cardamonin on TAMs. With the conditioned medium of ovarian cancer cells, macrophages were induced to TAMs and, accordingly, promoted the proliferation, migration, and invasion of ovarian cancer cells. Cardamonin suppressed alternatively activated (M2) polarization of TAMs and downregulated TAM-secreted tumorigenic factors, thereby hindering the pro-tumor function of TAMs on ovarian cancer cells. Moreover, cardamonin inhibited tumor growth in xenograft nude mice and lowered the expression of CD163 and CD206. Mechanistically, cardamonin inhibited the phosphorylation of mammalian target of rapamycin (mTOR) and signal transducer and activator of transcription 3 (STAT3), resulting in the suppression of M2 polarization. Furthermore, STAT3 is tightly related with mTOR activity. Altogether, these findings implied that cardamonin suppresses the pro-tumor function of TAMs by decreasing M2 polarization via mTOR inhibition, and cardamonin may be a potential therapeutic agent for ovarian cancer.
Collapse
Affiliation(s)
- Huajiao Chen
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, Fujian 350001, PR China
| | - Sheng Huang
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, PR China
| | - Peiguang Niu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, Fujian 350001, PR China
| | - Yanting Zhu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, Fujian 350001, PR China
| | - Jintuo Zhou
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, Fujian 350001, PR China
| | - Li Jiang
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, Fujian 350001, PR China
| | - Danyun Li
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, Fujian 350001, PR China
| | - Daohua Shi
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, Fujian 350001, PR China
| |
Collapse
|
84
|
Zhao L, Xu TS, Gong J, Cai YC. Survival and prognostic factors of ultra-low rectal cancer patients after anus-preserving surgery. Shijie Huaren Xiaohua Zazhi 2022; 30:762-768. [DOI: 10.11569/wcjd.v30.i17.762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Colorectal cancer is a highly malignant tumor. Anus-preserving surgery can effectively prolong the survival of patients with ultra-low rectal cancer. However, the postoperative survival of patients is affected by many factors. Identification of the relevant prognostic factors is of important guiding value in enhancing postoperative intervention and improving the quality of life of patients.
AIM To explore the survival of patients with ultra-low rectal cancer after anus-preserving surgery, and to analyze the prognostic factors.
METHODS A total of 152 patients with low rectal cancer were selected from March 2016 to March 2018 at our hospital, all of whom received anus-preserving surgery for ultra-low rectal cancer. The overall survival rate and disease-free survival rate at 1, 2, and 3 years after surgery were analyzed. The perioperative clinical data of patients with different prognostic conditions were compared, and the prognostic factors were analyzed.
RESULTS The 1-, 2-, and 3-year overall survival rates were 100%, 89.8% and 70.75%, respectively, and the 1-, 2-, and 3-year disease-free survival rates were 91.16%, 76.19%, and 60.54%, respectively. Age at surgery, tumor diameter, serum carbohydrate antigen 125 (CA125), carcinoembryonic antigen (CEA), hypoxia-inducing-factor 1α (HIF-1α) levels, TNM stage, and postoperative catheter indwelling time of patients who survived 3 years after surgery were all lower than those of patients who died, and the incidence of lymph node metastasis, vascular invasion, and nerve invasion was lower than that of patients who died. The distance from anal margin, serum miR-192 level, differentiation degree, and PNAG score were all significantly higher than those of dead patients (P < 0.05). The distance to the anal margin, serum CA125, CEA, HIF-1α, and miR-192 levels, PNAG score, and postoperative catheter indwelling time were significantly correlated with the prognosis of patients with ultra-low rectal cancer after anus-preserving surgery (P < 0.05).
CONCLUSION The 3-year overall survival rate of patients with ultra-low rectal cancer after anus-preserving surgery is more than 70%. The distance between tumor and anal margin, serum CA125, CEA, HIF-1α, and miR-192 levels, PNAG score, and postoperative catheter indwelling time are prognostic factors for these patients.
Collapse
Affiliation(s)
- Lei Zhao
- Quzhou Hospital Affiliated to Wenzhou Medical University (Quzhou People's Hospital), Quzhou 324000, Zhejiang Province, China
| | - Tian-Sheng Xu
- Quzhou Hospital Affiliated to Wenzhou Medical University (Quzhou People's Hospital), Quzhou 324000, Zhejiang Province, China
| | - Jiang Gong
- Quzhou Hospital Affiliated to Wenzhou Medical University (Quzhou People's Hospital), Quzhou 324000, Zhejiang Province, China
| | - Ying-Chang Cai
- Quzhou Hospital Affiliated to Wenzhou Medical University (Quzhou People's Hospital), Quzhou 324000, Zhejiang Province, China
| |
Collapse
|
85
|
Wang Y, He H, Chen J, Song Z, Pan X, Lan T, Wang G. Effects of glycolysis and polyamine predation on intestinal epithelial barrier in colorectal cancer. Front Oncol 2022; 12:961257. [PMID: 35912204 PMCID: PMC9337861 DOI: 10.3389/fonc.2022.961257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 06/24/2022] [Indexed: 01/15/2023] Open
Abstract
Colorectal cancer (CRC) is the second most lethal cancer and the third most common cancer in the world, and its prognosis is severely affected by high intestinal mucosal permeability and increasing tumor burden. Studies have shown that the expression of hypoxia induce factor 1α (HIF1α) is up-regulated in a variety of tumor tissues, which is related to multiple metabolic reprogramming of tumor cells. However, the role of HIF1α in CRC tumor growth, tumor polyamine metabolism and intestinal mucosal barrier damage has not been studied. Here, we constructed different types of CRC tumor-bearing mice models by inoculating HCT116 cells with different levels of HIF1α expression (knockdown, wild type, overexpression) in the armpits of mice to explore the upstream and downstream regulators of HIF1α, the effects of HIF1α on the growth of CRC, and the CRC polyamine metabolism and its effect on the intestinal mucosal barrier. We found that with the increase of HIF1 gene expression, tumor growth was promoted and intestinal mucosal permeability was increased. The expression of glycolysis-related proteins was up-regulated, the rate-limiting enzyme ODC of polyamine synthesis was decreased, and the transfer protein of polyamine was increased. HPLC showed that the polyamine content in the tumor tissue of the overexpression group HIF1α OE was higher than that of the wild group HIF1α (+/+), and higher than that of the knockdown group HIF1α (-/-), but the content of polyamines in intestinal mucosa was the opposite. After supplementation of exogenous polyamines, the content of polyamines in intestinal mucosa and tumor tissue increased, and the damage of intestinal mucosa was alleviated. In conclusion, upon activation of the MYC/HIF1 pathway, tumor glycolysis is enhanced, tumors require more energy and endogenous polyamine synthesis is reduced. Therefore, in order to meet its growth needs, tumor will rob polyamines in the intestinal mucosa, resulting in intestinal mucosal epithelial barrier dysfunction.
Collapse
Affiliation(s)
| | | | | | | | | | - Tian Lan
- *Correspondence: Tian Lan, ; Guixiang Wang,
| | | |
Collapse
|
86
|
Huang Y, Du Y, Zheng Y, Wen C, Zou H, Huang J, Zhou H, Zhao H, Wu L. Ct-OATP1B3 promotes high-grade serous ovarian cancer metastasis by regulation of fatty acid beta-oxidation and oxidative phosphorylation. Cell Death Dis 2022; 13:556. [PMID: 35717493 PMCID: PMC9206684 DOI: 10.1038/s41419-022-05014-1] [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: 09/01/2021] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 01/21/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most lethal gynecologic malignancy mainly due to its extensive metastasis. Cancer-type organic anion transporting polypeptide 1B3 (Ct-OATP1B3), a newly discovered splice variant of solute carrier organic anion transporter family member 1B3 (SLCO1B3), has been reported to be overexpressed in several types of cancer. However, the biological function of Ct-OATP1B3 remains largely unknown. Here, we reveal that Ct-OATP1B3 is overexpressed in HGSOC and promotes the metastasis of HGSOC in vivo and in vitro. Mechanically, Ct-OATP1B3 directly interacts with insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2), an RNA-binding protein, which results in enhancement of the mRNA stability and expression of carnitine palmitoyltransferase 1A (CPT1A) and NADH:Ubiquinone Oxidoreductase Subunit A2 (NDUFA2), leading to increased mitochondrial fatty acid beta-oxidation (FAO) and oxidative phosphorylation (OXPHOS) activities. The increased FAO and OXPHOS activities further facilitate adenosine triphosphate (ATP) production and cellular lamellipodia formation, which is the initial step in the processes of tumor cell migration and invasion. Taken together, our study provides an insight into the function and underlying mechanism of Ct-OATP1B3 in HGSOC metastasis, and highlights Ct-OATP1B3 as a novel prognostic marker as well as therapeutic target in HGSOC.
Collapse
Affiliation(s)
- Yutang Huang
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 China
| | - Yan Du
- grid.8547.e0000 0001 0125 2443Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China ,grid.8547.e0000 0001 0125 2443Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, 200032 China
| | - Yujie Zheng
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 China
| | - Chunjie Wen
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 China
| | - Hecun Zou
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 China
| | - Jiafeng Huang
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 China
| | - Honghao Zhou
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 China ,grid.216417.70000 0001 0379 7164Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Central South University, Changsha, 410078 China
| | - Hongbo Zhao
- grid.8547.e0000 0001 0125 2443Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China ,grid.8547.e0000 0001 0125 2443Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, 200032 China ,grid.412312.70000 0004 1755 1415Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011 China
| | - Lanxiang Wu
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 China ,grid.203458.80000 0000 8653 0555Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, 400016 China
| |
Collapse
|
87
|
Blevins HM, Xu Y, Biby S, Zhang S. The NLRP3 Inflammasome Pathway: A Review of Mechanisms and Inhibitors for the Treatment of Inflammatory Diseases. Front Aging Neurosci 2022; 14:879021. [PMID: 35754962 PMCID: PMC9226403 DOI: 10.3389/fnagi.2022.879021] [Citation(s) in RCA: 147] [Impact Index Per Article: 73.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/12/2022] [Indexed: 12/24/2022] Open
Abstract
The NLRP3 inflammasome is a multiprotein complex that plays a pivotal role in regulating the innate immune system and inflammatory signaling. Upon activation by PAMPs and DAMPs, NLRP3 oligomerizes and activates caspase-1 which initiates the processing and release of pro-inflammatory cytokines IL-1β and IL-18. NLRP3 is the most extensively studied inflammasome to date due to its array of activators and aberrant activation in several inflammatory diseases. Studies using small molecules and biologics targeting the NLRP3 inflammasome pathway have shown positive outcomes in treating various disease pathologies by blocking chronic inflammation. In this review, we discuss the recent advances in understanding the NLRP3 mechanism, its role in disease pathology, and provide a broad review of therapeutics discovered to target the NLRP3 pathway and their challenges.
Collapse
Affiliation(s)
| | | | | | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
88
|
Ma W, Sun X, Zhang S, Chen Z, Yu J. Circ_0039960 regulates growth and Warburg effect of breast cancer cells via modulating miR-1178/PRMT7 axis. Mol Cell Probes 2022; 64:101829. [PMID: 35597500 DOI: 10.1016/j.mcp.2022.101829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Breast cancer (BC) is a serious threat to women's life and healthy. Increasing evidence indicated that blocking Warburg effect could attenuate the development of BC. Circular RNAs (circRNAs) has been found to be dysregulated in various carcinomas, including BC. Our study aims to illustrate the role and regulatory mechanism of circ_0039960 in BC development. METHODS RT-qPCR and western blotting were utilized to evaluate the expression of circ_0039960 in tissues recruited from 32 cases of BC patients and also BC cell lines. Circ_0039960 shRNA was transfected into cells to explore its function on cell processes. CCK-8, flow cytometry and ELISA were used to measure cell viability, cell cycle and apoptosis. Warburg effect was detected by using commercial kits. Besides, bioinformatic prediction, RIP and luciferase reporter assays were performed to validate the interactions between circ_0039960, miR-1178 and PRMT7. RESULTS The results showed that circ_0039960 and PRMT7 were both up-regulated, while miR-1178 was down-regulated, in BC tissues and cells. Silencing circ_0039960 effectively inhibited cell viability and Warburg effect of BC cells, also, induced cell cycle arrest and apoptosis. Moreover, we validated that circ_0039960 positively mediated PRMT7 expression via directly targeting to miR-1178. The inhibition of miR-1178 and overexpression of PRMT7 reversed the effect of circ_0039960 knockdown on BC cell growth and Warburg effect. CONCLUSION In general, our research demonstrated that circ_0039960 regulates cell growth and Warburg effect in BC cells via miR-1178/PRMT7 axis. This may provide new evidence for the exploration of BC diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Weichang Ma
- Department of Thyroid and Breast Surgury, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong province, China
| | - Xiaojun Sun
- Department of Thyroid and Breast Surgury, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong province, China
| | - Shupeng Zhang
- Intensive Care Unit, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong province, China
| | - Zhenghua Chen
- Department of Thyroid and Breast Surgury, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong province, China
| | - Jianing Yu
- Department of Thyroid and Breast Surgury, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong province, China.
| |
Collapse
|
89
|
Wiggs A, Molina S, Sumner SJ, Rushing BR. A Review of Metabolic Targets of Anticancer Nutrients and Nutraceuticals in Pre-Clinical Models of Triple-Negative Breast Cancer. Nutrients 2022; 14:1990. [PMID: 35631131 PMCID: PMC9146055 DOI: 10.3390/nu14101990] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/19/2022] [Accepted: 05/06/2022] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is notoriously aggressive and has poorer outcomes as compared with other breast cancer subtypes. Due to a lack of targeted therapies, TNBC is often treated with chemotherapeutics as opposed to hormone therapy or other targeted therapies available to individuals with estrogen receptor positive (ER+) breast cancers. Because of the lack of treatment options for TNBC, other therapeutic avenues are being explored. Metabolic reprogramming, a hallmark of cancer, provides potential opportunities to target cancer cells more specifically, increasing efficacy and reducing side effects. Nutrients serve a significant role in metabolic processes involved in DNA transcription, protein folding, and function as co-factors in enzyme activity, and may provide novel strategies to target cancer cell metabolism in TNBC. This article reviews studies that have investigated how nutrients/nutraceuticals target metabolic processes in TNBC cells alone or in combination with existing drugs to exert anticancer effects. These agents have been shown to cause perturbations in many metabolic processes related to glucose metabolism, fatty acid metabolism, as well as autophagy and oxidative stress-related metabolism. With this information, we present the potential of nutrients as metabolism-directed anticancer agents and the potential for using these agents alone or in cocktails as a new direction for TNBC therapy.
Collapse
Affiliation(s)
- Alleigh Wiggs
- Department of Nutrition, University of North Carolina-Chapel Hill, Durham, NC 27599, USA
| | - Sabrina Molina
- Nutrition Research Institute, University of North Carolina-Chapel Hill, Kannapolis, NC 280821, USA
| | - Susan J. Sumner
- Department of Nutrition, University of North Carolina-Chapel Hill, Durham, NC 27599, USA
- Nutrition Research Institute, University of North Carolina-Chapel Hill, Kannapolis, NC 280821, USA
| | - Blake R. Rushing
- Department of Nutrition, University of North Carolina-Chapel Hill, Durham, NC 27599, USA
- Nutrition Research Institute, University of North Carolina-Chapel Hill, Kannapolis, NC 280821, USA
| |
Collapse
|
90
|
Jiang Y, Zhang M, Yu D, Hou G, Wu J, Li F. CircRBM33 downregulation inhibits hypoxia-induced glycolysis and promotes apoptosis of breast cancer cells via a microRNA-542-3p/HIF-1α axis. Cell Death Dis 2022; 8:126. [PMID: 35318311 PMCID: PMC8941146 DOI: 10.1038/s41420-022-00860-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 01/06/2022] [Accepted: 01/20/2022] [Indexed: 11/14/2022]
Abstract
Many circRNAs are involved in the carcinogenesis of breast cancer (BCa) through the transcription of microRNAs (miRNAs) and mRNAs. This study investigated circRBM33 regulation of the miR-542-3p/hypoxia-inducible factor-1α (HIF-1α) axis in BCa. BCa clinical tissue samples were collected to test differential expressions of circRBM33, miR-542-3p, and HIF-1α. MCF-7 cells were subjected to normoxia or hypoxia and transfected with plasmids that regulated CircRBM33, miR-542-3p, and HIF-1α expression levels. Glycolysis was evaluated by measuring glucose consumption, lactic acid production, and protein expression of hexokinase 2, glucose transporter type 1 and lactic dehydrogenase A. Cell proliferation and apoptosis were also assessed, and the interactions between genes were explored. CircRBM33 and HIF-1α were upregulated, while miR-542-3p was downregulated in BCa tissue samples and cell lines. Hypoxia induced circRBM33 expression in BCa cells, which negatively regulated miR-542-3p expression. CircRBM33 knockdown or miR-542-3p rescue reduced glycolysis and proliferation and promoted apoptosis of BCa cells. MiR-542-3p inhibition rescued circRBM33 knockdown-mediated glycolysis, proliferation and apoptosis of BCa cells. MiR-542-3p targeted HIF-1α, and the overexpression of HIF-1α reversed the effect of miR-542-3p upregulation on glycolysis, proliferation, and apoptosis of BCa cells. Collectively, downregulating circRBM33 suppresses miR-542-3p-targeted HIF-1α expression, resulting in the inhibition of glycolysis and proliferation and the promotion of BCa cells’ apoptosis.
Collapse
Affiliation(s)
- Yiming Jiang
- Department of Oncology, Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang Province, China
| | - Meiqi Zhang
- Department of Outpatient, Affiliated Hospital of Jiaxing University, Jiaxing, 314001, Zhejiang Province, China
| | - Danlu Yu
- Department of Endocrinology, Affiliated Hospital of Jiaxing University, Jiaxing, 314001, Zhejiang Province, China
| | - Guoxin Hou
- Department of Oncology, Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang Province, China
| | - Jingyi Wu
- Department of Oncology, Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang Province, China
| | - Fuming Li
- Department of Breast Surgery, Second Hospital of Jiaxing City, Jiaxing, 314000, Zhejiang Province, China.
| |
Collapse
|
91
|
Wang T, Tian L, Cheng Q, Feng S, Zhang H, Zheng Z, Liu Y, Cheng M, Meng Z, Meng Q. Pep5-based antitumor peptides containing multifunctional fragments with enhanced activity and synergistic effect. Eur J Med Chem 2022; 237:114320. [DOI: 10.1016/j.ejmech.2022.114320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 12/27/2022]
|
92
|
Cardamomin protects from diabetes-induced kidney damage through modulating PI3K/AKT and JAK/STAT signaling pathways in rats. Int Immunopharmacol 2022; 107:108610. [PMID: 35219163 DOI: 10.1016/j.intimp.2022.108610] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/24/2022] [Accepted: 02/03/2022] [Indexed: 12/19/2022]
Abstract
BACKGROUND Diabetic nephropathy is one of the common complications of diabetes mellitus, which seriously affects the life quality and health of patients. In this study, we aimed to investigate the function of cardamonin (CAD) in diabetes-induced kidney damage in rats. METHODS The normal rat kidney tubular epithelial cells (NRK-52E) were pre-treated with different doses of CAD and then stimulated with methylglyoxal (MGO). Streptozotocin (STZ) induced diabetes rat model were received different doses of CAD treatment. MTT, EdU, Transwell, and flow cytometry was used to detect cell viability, proliferation, migration, and apoptosis. Western blot analysis was used to detect the expression of apoptosis related proteins, advanced glycation end-products (AGEs), receptor for AGEs (RAGE), epithelial mesenchymal transition (EMT) related proteins, phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway related proteins, and janus kinas/signal transducer and activator of transcription 3 (JAK/STAT3) related proteins. ELISA assay was used to detect the levels of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β). The levels of malondialdehyde (MDA), glutathione (GSH), and superoxide dismutase (SOD) were detected using commercial kit. Hematoxylin and eosin staining was used to assess pathological changes in rat kidney. RESULTS Compared with control group, MGO reduced cell viability and proliferation, enhanced migration and apoptosis of NRK-52E cells, while CAD inhibited these effects induced by MGO in NRK-52E cells. Moreover, CAD increased Bcl-2 expression and decreased the expression of Bax and cleaved caspase-3 in MGO-treated NRK-52E cells. Compared with control group, MGO increased the AGEs formation, the expression of RAGE and p-p65, the levels of TNF-α, IL-6, IL-1β, MDA in NRK-52E cells and reduced the levels of GSH and SOD, while treatment of CAD dose-dependently prevented these results. In addition, CAD attenuated MGO-induced EMT of MGO-treated NRK-52E cells. Mechanically, we identified that CAD repressed PI3K/AKT and JAK/STAT3 signaling in NRK-52E cells. Importantly, the kidney injury of diabetes rats was attenuated by CAD. Besides, STZ-induced inflammatory response, oxidative stress, and phosphorylation levels of PI3K, AKT, JAK2, and STAT3 were reduced by CAD in the rats. CONCLUSION CAD protects from diabetes-induced kidney damage through modulating PI3K/AKT and JAK/STAT signaling pathways in rats.
Collapse
|
93
|
Bouhamida E, Morciano G, Perrone M, Kahsay AE, Della Sala M, Wieckowski MR, Fiorica F, Pinton P, Giorgi C, Patergnani S. The Interplay of Hypoxia Signaling on Mitochondrial Dysfunction and Inflammation in Cardiovascular Diseases and Cancer: From Molecular Mechanisms to Therapeutic Approaches. BIOLOGY 2022; 11:biology11020300. [PMID: 35205167 PMCID: PMC8869508 DOI: 10.3390/biology11020300] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary The regulation of hypoxia has recently emerged as having a central impact in mitochondrial function and dysfunction in various diseases, including the major disorders threatening worldwide: cardiovascular diseases and cancer. Despite the studies in this matter, its effective role in protection and disease progression even though its direct molecular mechanism in both disorders is still to be elucidated. This review aims to cover the current knowledge about the effect of hypoxia on mitochondrial function and dysfunction, and inflammation, in cardiovascular diseases and cancer, and reports further therapeutic strategies based on the modulation of hypoxic pathways. Abstract Cardiovascular diseases (CVDs) and cancer continue to be the primary cause of mortality worldwide and their pathomechanisms are a complex and multifactorial process. Insufficient oxygen availability (hypoxia) plays critical roles in the pathogenesis of both CVDs and cancer diseases, and hypoxia-inducible factor 1 (HIF-1), the main sensor of hypoxia, acts as a central regulator of multiple target genes in the human body. Accumulating evidence demonstrates that mitochondria are the major target of hypoxic injury, the most common source of reactive oxygen species during hypoxia and key elements for inflammation regulation during the development of both CVDs and cancer. Taken together, observations propose that hypoxia, mitochondrial abnormality, oxidative stress, inflammation in CVDs, and cancer are closely linked. Based upon these facts, this review aims to deeply discuss these intimate relationships and to summarize current significant findings corroborating the molecular mechanisms and potential therapies involved in hypoxia and mitochondrial dysfunction in CVDs and cancer.
Collapse
Affiliation(s)
- Esmaa Bouhamida
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48022 Cotignola, Italy
| | - Giampaolo Morciano
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48022 Cotignola, Italy
| | - Mariasole Perrone
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
| | - Asrat E. Kahsay
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
| | - Mario Della Sala
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02-093 Warsaw, Poland;
| | - Francesco Fiorica
- Department of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, Ospedale Mater Salutis di Legnago, 37045 Verona, Italy;
| | - Paolo Pinton
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48022 Cotignola, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
- Correspondence: (C.G.); (S.P.)
| | - Simone Patergnani
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48022 Cotignola, Italy
- Correspondence: (C.G.); (S.P.)
| |
Collapse
|
94
|
Ni H, Li J, Zheng J, Zhou B. Cardamonin attenuates cerebral ischemia/reperfusion injury by activating the HIF-1α/VEGFA pathway. Phytother Res 2022; 36:1736-1747. [PMID: 35142404 DOI: 10.1002/ptr.7409] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 01/17/2022] [Accepted: 01/27/2022] [Indexed: 12/13/2022]
Abstract
Cardamonin is a chalcone with neuroprotective activity. The aim of our study was to explore the functions and mechanism of action of cardamonin in ischemic stroke. Oxygen-glucose deprivation and reperfusion (OGD/R)-induced human brain microvascular endothelial cells (HBMECs) and middle cerebral artery occlusion (MCAO) mouse model were utilized to mimic ischemic stroke. Cell viability was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide. Permeability was investigated via fluorescein isothiocyanate-dextran assay. Apoptosis was detected by TdT-Mediated dUTP Nick End Labeling staining. Hypoxia-inducible factor (HIF)-1α and vascular endothelial growth factor A (VEGFA) protein levels were measured using Western blotting. Brain injury was evaluated by 2,3,5-triphenyltetrazolium chloride staining, neurological score and brain water content. The 37 overlapping targets of ischemic stroke and cardamonin were predicted to be associated with the HIF-1/VEGFA signaling. Cardamonin alleviated OGD/R-induced viability reduction and increase of permeability and apoptosis in HBMECs. Cardamonin increased OGD/R-induced activation of the HIF-1α/VEGFA pathway. Inhibition of the HIF-1α/VEGFA signaling using inhibitor relieved the effect of cardamonin on cell viability, permeability and apoptosis in HBMECs under OGD/R. Cardamonin mitigated brain injury and promoted activation of the HIF-1α/VEGFA signaling in MCAO-treated mice. Overall, cardamonin protected against OGD/R-induced HBMEC damage and MACO-induced brain injury through activating the HIF-1α/VEGFA pathway.
Collapse
Affiliation(s)
- Hongzao Ni
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, China
| | - Jinxiao Li
- Department of Neurosurgery, Xinyi People's Hospital, Xuzhou, China
| | - Jinyu Zheng
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, China
| | - Botao Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
95
|
Zhu X, Zhang Y, Wang Y, Zhang H, Wang X, Tang H, Huang H, Zhou Z, Chen B, Sun L. Agrimoniin sensitizes pancreatic cancer to apoptosis through ROS-mediated energy metabolism dysfunction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 96:153807. [PMID: 34785107 DOI: 10.1016/j.phymed.2021.153807] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/09/2021] [Accepted: 10/17/2021] [Indexed: 05/19/2023]
Abstract
BACKGROUND Pancreatic cancer is a fatal tumor, which is one of the most common malignant tumors at present. Patients with pancreatic cancer also respond poorly to chemotherapy or radiation therapy and may be accompanied by serious adverse reactions. Therefore, to find an effective way to inhibit the initiation and progression of pancreatic cancer is important to improve the survival and development of patients. Agrimoniin, a polyphenol compounds isolated from Agrimonia pilosa ledeb, has antiviral, antimicrobial, and anticancer activities in vivo and in vitro. However, its molecular mechanism in pancreatic cancer remains to be determined. PURPOSE We aimed to investigate the effect of agrimoniin in pancreatic cancer and its underlying mechanism in vivo and in vitro. METHODS The proliferation was detected by colony formation, cell proliferation and toxicity, and real-time cell analysis techniques. The apoptosis was detected by flow cytometry and Western blot. Flow cytometry was used to measure the level of reactive oxygen species (ROS) and apoptosis. The level of intracellular ROS or mitochondrial membrane potential was measured with a DCFH-DA or JC-1 probe. Cell metabolism assays were analyzed and evaluated by using Agilent Seahorse Bioscience XF96 Extracellular Flux Analyzer. The target proteins were analyzed by Western blot. Subcutaneous cancer models in nude mice were established to evaluate the anticancer effects in vivo. RESULTS Agrimoniin inhibited cell growth and promoted cell apoptosis by regulating cell metabolism in pancreatic cancer cells. Agrimoniin increased the ROS level in pancreatic cancer cells by suppressing Nrf2-dependent ROS scavenging system and disrupting normal mitochondrial membrane potential. We also found that agrimoniin significantly disrupted mitochondrial function and reduced the protein expression of mTOR/HIF-1α pathway and subsequently decreased oxygen consumption rate and extracellular acidification rate. Eventually, agrimoniin affected intracellular energy metabolism and induced apoptosis of pancreatic cancer cells. CONCLUSIONS These findings reveal the novel function of agrimoniin in promoting apoptosis of pancreatic cancer cells through mediating energy metabolism dysfunction. Altogether, the potential new targets and their synergies discovered in this research are of great significance for cancer treatment and drug development.
Collapse
Affiliation(s)
- Xiandong Zhu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yan Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yongqiang Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Hewei Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Xiaowu Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Hengjie Tang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Hongjian Huang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Zijun Zhou
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Bicheng Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China.
| | - Linxiao Sun
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China.
| |
Collapse
|
96
|
Pan T, Sun S, Chen Y, Tian R, Chen E, Tan R, Wang X, Liu Z, Liu J, Qu H. Immune effects of PI3K/Akt/HIF-1α-regulated glycolysis in polymorphonuclear neutrophils during sepsis. Crit Care 2022; 26:29. [PMID: 35090526 PMCID: PMC8796568 DOI: 10.1186/s13054-022-03893-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/10/2022] [Indexed: 12/29/2022] Open
Abstract
Background Effective removal of pathogenic bacteria is key to improving the prognosis of sepsis. Polymorphonuclear neutrophils (PMNs) are the most important components of innate cellular immunity and play vital roles in clearing pathogenic bacteria. However, the metabolic characteristics and immunomodulatory pathways of PMNs during sepsis have not been investigated. In the present study, we explored the immune metabolism characteristics of PMNs and the mechanism by which neutrophilic glycolysis is regulated during sepsis. Methods Metabolomics analysis was performed on PMNs isolated from 14 septic patients, 26 patients with acute appendicitis, and 19 healthy volunteers. Transcriptome analysis was performed on the PMNs isolated from the healthy volunteers and the patients with sepsis to assess glycolysis and investigate its mechanism. Lipopolysaccharide (LPS) was used to stimulate the neutrophils isolated from the healthy volunteers at different time intervals to build an LPS-tolerant model. Chemotaxis, phagocytosis, lactate production, oxygen consumption rate (OCR), and extracellular acidification rate (ECAR) were evaluated. Results Transcriptomics showed significant changes in glycolysis and the mTOR/HIF-1α signaling pathway during sepsis. Metabolomics revealed that the Warburg effect was significantly altered in the patients with sepsis. We discovered that glycolysis regulated PMNs’ chemotaxis and phagocytosis functions during sepsis. Lactate dehydrogenase A (LDHA) downregulation was a key factor in the inhibition of glycolysis in PMNs. This study confirmed that the PI3K/Akt-HIF-1α pathway was involved in the LDHA expression level and also influenced PMNs’ chemotaxis and phagocytosis functions. Conclusions The inhibition of glycolysis contributed to neutrophil immunosuppression during sepsis and might be controlled by PI3K/Akt-HIF-1α pathway-mediated LDHA downregulation. Our study provides a scientific theoretical basis for the management and treatment of patients with sepsis and promotes to identify therapeutic target for the improvement of immune function in sepsis. Supplementary Information The online version contains supplementary material available at 10.1186/s13054-022-03893-6.
Collapse
|
97
|
Sun H, Zhang N, Jin Y, Xu H. Cardamonin Promotes the Apoptosis and Chemotherapy Sensitivity to Gemcitabine of Pancreatic Cancer Through Modulating the FOXO3a-FOXM1 Axis. Dose Response 2022; 19:15593258211042163. [PMID: 34987330 PMCID: PMC8725235 DOI: 10.1177/15593258211042163] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022] Open
Abstract
Cardamonin (CAR), a flavone existing in the Alpinia plant, has been found to
modulate multiple biological activities, including antioxidant,
anti-inflammatory, and anti-tumor effects. Nevertheless, the influence of CAR on
pancreatic cancer (PC) is less understood. Here, we conducted in
vitro and in vivo experiments to explore the
functions of CAR on PC cells’ proliferation, apoptosis and chemosensitivity to
gemcitabine (GEM). The growth of PC cells (including PANC-1 and SW1990) was
evaluated by the cell counting kit-8 assay, colony formation assay and xenograft
tumor experiment. Besides, the apoptosis was determined by flow cytometry and
western blot (WB). Moreover, the FOXO3a-FOXM1 pathway expression was tested by
reverse transcription-polymerase chain reaction and WB. Our data suggested that
CAR restrained cell proliferation, growth and expedited apoptosis both
in vitro and in vivo. Moreover, CAR
sensitized PC cells to GEM. Mechanistically, CAR heightened FOXO3a while
repressed FOXM1. Further loss-of-function assays revealed that down-regulating
FOXO3a markedly dampened the anti-tumor effect induced by CAR and accelerated
the FOXM1 expression. Our data confirmed that CAR exerted an anti-tumor function
in PC dependently by modulating the FOXO3a-FOXM1 axis.
Collapse
Affiliation(s)
- Huapeng Sun
- Department of General Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Na Zhang
- Department of Pathology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Yiqiang Jin
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Haisheng Xu
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| |
Collapse
|
98
|
Mahmoud AM, Sayed AM, Ahmed OS, Abdel-Daim MM, Hassanein EHM. The role of flavonoids in inhibiting IL-6 and inflammatory arthritis. Curr Top Med Chem 2022; 22:746-768. [PMID: 34994311 DOI: 10.2174/1568026622666220107105233] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/21/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that primarily affects the synovial joints. RA has well-known clinical manifestations and can cause progressive disability and premature death along with socioeconomic burdens. Interleukin-6 (IL-6) has been implicated in the pathology of RA where it can stimulate pannus formation, osteoclastogenesis, and oxidative stress. Flavonoids are plant metabolites with beneficial pharmacological effects, including anti-inflammatory, antioxidant, antidiabetic, anticancer, and others. Flavonoids are polyphenolic compounds found in a variety of plants, vegetables, and fruits. Many flavonoids have demonstrated anti-arthritic activity mediated mainly through the suppression of pro-inflammatory cytokines. This review thoroughly discusses the accumulate data on the role of flavonoids on IL-6 in RA.
Collapse
Affiliation(s)
- Ayman M Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Egypt
| | - Osama S Ahmed
- Faculty of Pharmacy, Al-Azhar University-Assiut Branch, Egypt
| | - Mohamed M Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University-Assiut Branch, Egypt
| |
Collapse
|
99
|
Shen LW, Jiang XX, Li ZQ, Li J, Wang M, Jia GF, Ding X, Lei L, Gong QH, Gao N. Cepharanthine sensitizes human triple negative breast cancer cells to chemotherapeutic agent epirubicin via inducing cofilin oxidation-mediated mitochondrial fission and apoptosis. Acta Pharmacol Sin 2022; 43:177-193. [PMID: 34294886 PMCID: PMC8724299 DOI: 10.1038/s41401-021-00715-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
Inhibition of autophagy has been accepted as a promising therapeutic strategy in cancer, but its clinical application is hindered by lack of effective and specific autophagy inhibitors. We previously identified cepharanthine (CEP) as a novel autophagy inhibitor, which inhibited autophagy/mitophagy through blockage of autophagosome-lysosome fusion in human breast cancer cells. In this study we investigated whether and how inhibition of autophagy/mitophagy by cepharanthine affected the efficacy of chemotherapeutic agent epirubicin in triple negative breast cancer (TNBC) cells in vitro and in vivo. In human breast cancer MDA-MB-231 and BT549 cells, application of CEP (2 μM) greatly enhanced cepharanthine-induced inhibition on cell viability and colony formation. CEP interacted with epirubicin synergistically to induce apoptosis in TNBC cells via the mitochondrial pathway. We demonstrated that co-administration of CEP and epirubicin induced mitochondrial fission in MDA-MB-231 cells, and the production of mitochondrial superoxide was correlated with mitochondrial fission and apoptosis induced by the combination. Moreover, we revealed that co-administration of CEP and epirubicin markedly increased the generation of mitochondrial superoxide, resulting in oxidation of the actin-remodeling protein cofilin, which promoted formation of an intramolecular disulfide bridge between Cys39 and Cys80 as well as Ser3 dephosphorylation, leading to mitochondria translocation of cofilin, thus causing mitochondrial fission and apoptosis. Finally, in mice bearing MDA-MB-231 cell xenografts, co-administration of CEP (12 mg/kg, ip, once every other day for 36 days) greatly enhanced the therapeutic efficacy of epirubicin (2 mg/kg) as compared with administration of either drug alone. Taken together, our results implicate that a combination of cepharanthine with chemotherapeutic agents could represent a novel therapeutic strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Li-wen Shen
- grid.417409.f0000 0001 0240 6969Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006 China
| | - Xiu-xing Jiang
- grid.410570.70000 0004 1760 6682College of Pharmacy, Army Medical University, Chongqing, 400038 China
| | - Zhi-qiang Li
- grid.410570.70000 0004 1760 6682College of Pharmacy, Army Medical University, Chongqing, 400038 China
| | - Jie Li
- grid.410570.70000 0004 1760 6682College of Pharmacy, Army Medical University, Chongqing, 400038 China
| | - Mei Wang
- grid.417409.f0000 0001 0240 6969Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006 China
| | - Guan-fei Jia
- grid.410570.70000 0004 1760 6682College of Pharmacy, Army Medical University, Chongqing, 400038 China
| | - Xin Ding
- grid.410570.70000 0004 1760 6682College of Pharmacy, Army Medical University, Chongqing, 400038 China
| | - Ling Lei
- grid.410570.70000 0004 1760 6682College of Pharmacy, Army Medical University, Chongqing, 400038 China
| | - Qi-hai Gong
- grid.417409.f0000 0001 0240 6969Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006 China
| | - Ning Gao
- grid.417409.f0000 0001 0240 6969Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006 China ,grid.410570.70000 0004 1760 6682College of Pharmacy, Army Medical University, Chongqing, 400038 China
| |
Collapse
|
100
|
Wen X, Zhang L, Zhao S, Liu Q, Guan W, Wu J, Zhang Q, Wen H, Huang W. High-Throughput Screening and Identification of Human Adenovirus Type 5 Inhibitors. Front Cell Infect Microbiol 2021; 11:767578. [PMID: 34976856 PMCID: PMC8718806 DOI: 10.3389/fcimb.2021.767578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/29/2021] [Indexed: 12/02/2022] Open
Abstract
Human adenovirus infections can develop into diffuse multi-organ diseases in young children and immunocompromised patients, and severe cases can lead to death. However, there are no approved antiviral drugs available to treat adenovirus diseases. In this study, a chemiluminescence-based, high-throughput screening (HTS) assay was developed and applied to screen human adenovirus 5(HAdV5)inhibitors from 1,813 approved drug library and 556 traditional Chinese medicine-sourced small-molecule compounds. We identified three compounds with in vitro anti-HAdV5 activities in the low-micromolar range (EC50 values 0.3-4.5 μM, selectivity index values 20-300) that also showed inhibitory effects on HAdV3. Cardamomin (CDM) had good anti-HAdV5 activity in vitro. Furthermore, three dilutions of CDM (150, 75, and 37.5 mg/kg/d) administered to BALB/c mouse models inhibited HAdV5-fluc infection at 1 day post-infection by 80% (p < 0.05), 76% (p < 0.05), and 58% (p < 0.05), respectively. HE-staining of pathological tissue sections of mice infected with a wildtype adenoviral strain showed that CDM had a protective effect on tissues, especially in the liver, and greatly inhibited virus-induced necrosis of liver tissue. Thus, CDM inhibits adenovirus replication in vivo and in vitro. This study established a high-throughput screening method for anti-HAdV5 drugs and demonstrated CDM to be a candidate for HAdV5 therapy, potentially providing a new treatment for patients infected with adenoviruses.
Collapse
Affiliation(s)
- Xiaojing Wen
- Division of Human Immunodeficiency Virus (HIV)/Acquired Immune Deficiency Syndrome (AIDS) and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and World Health Organization (WHO) Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
- Department of Microbiological Laboratory Technology, School of Public Health, Cheeloo College of Medicine, Key Laboratory for the Prevention and Control of Infectious Diseases, Shandong University, Jinan, China
- Key Laboratory of China’s “13th Five-Year”, Shandong University, Jinan, China
| | - Li Zhang
- Division of Human Immunodeficiency Virus (HIV)/Acquired Immune Deficiency Syndrome (AIDS) and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and World Health Organization (WHO) Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Shan Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qiang Liu
- Division of Human Immunodeficiency Virus (HIV)/Acquired Immune Deficiency Syndrome (AIDS) and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and World Health Organization (WHO) Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Wenyi Guan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiajing Wu
- Division of Human Immunodeficiency Virus (HIV)/Acquired Immune Deficiency Syndrome (AIDS) and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and World Health Organization (WHO) Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Qiwei Zhang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Hongling Wen
- Department of Microbiological Laboratory Technology, School of Public Health, Cheeloo College of Medicine, Key Laboratory for the Prevention and Control of Infectious Diseases, Shandong University, Jinan, China
- Key Laboratory of China’s “13th Five-Year”, Shandong University, Jinan, China
| | - Weijin Huang
- Division of Human Immunodeficiency Virus (HIV)/Acquired Immune Deficiency Syndrome (AIDS) and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and World Health Organization (WHO) Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| |
Collapse
|