1
|
Xu Y, Chen L, Hu X, Lai Z, Chen B, Wu M, Mai L, Su Z, Chen J, Lai Z, Ai W, Xie J, Liao H, Xie Y. Brusatol ameliorates intestinal mucosal injury in ulcerative colitis via activating IL-22/STAT3 pathway. Int Immunopharmacol 2025; 153:114482. [PMID: 40101416 DOI: 10.1016/j.intimp.2025.114482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/23/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
Brusatol (BR) is an active compounds isolated from Brucea javanica, a Chinese herbal medicine that is famous for its anti-diarrheal effect. We have previously reported that BR mitigated inflammation in murine ulcerative colitis (UC) models. However, BR's role in intestinal mucosal healing, which is recently established as central strategy for the prevention and treatment of UC, remains unknown. In this study, the ameliorative effect of BR on intestinal mucosal damage was investigated in DSS-induced UC mice. BR significantly alleviated colitis symptoms, improved intestinal barrier function by preventing loss of goblet cells and downregulation of mucins and tight junction proteins, as well as maintained proliferative and apoptotic homeostasis in the colonic epithelium of UC mice. Mechanistically, BR enhanced the level and secretion of IL-22, but inhibited IL-22BP, an inhibitory protein of IL-22, in the blood serum and intestinal tissues of UC mice, as well as in MNK3 cells which is an effective cell model for studying ILC3s. Additionally, BR elevated the expressions of receptors for IL-22 (IL-10R2 and IL-22R1), and activated its downstream STAT3 signaling pathway. Furthermore, the involvement of IL-22 was further investigated by using recombinant IL-22 (rIL-22) and IL-22 antibody (anti-IL-22). BR demonstrated comparable effects with rIL-22 on alleviating intestinal inflammation and repairing intestinal mucosal injury. Treatment with anti-IL-22 abrogated the mucosal protective effects of BR. The present findings shed novel insights into the role of BR in intestinal mucosal healing via activating IL-22/STAT3 signaling pathway in UC.
Collapse
Affiliation(s)
- Ying Xu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Li Chen
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China; Pharmacy Center, Shenzhen Nanshan Medical Group Headquarters, Shenzhen, PR China
| | - Xiaoxia Hu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Zixuan Lai
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Baoyi Chen
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Minghui Wu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Liting Mai
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Ziren Su
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Jiannan Chen
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Zhengquan Lai
- Department of Pharmacy, Shenzhen University General Hospital/Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, PR China
| | - Weipeng Ai
- Department of Pharmacy, Shenzhen University General Hospital/Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, PR China
| | - Jianhui Xie
- Guangdong Provincial Key Laboratory of Clinical Research on TCM Syndrome, Guangzhou, PR China
| | - Huijun Liao
- Pharmacy Center, Shenzhen Nanshan Medical Group Headquarters, Shenzhen, PR China; Department of Clinical Pharmacy and Pharmaceutical Services, Shenzhen Nanshan People's Hospital, Shenzhen, PR China.
| | - Youliang Xie
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Clinical Research on TCM Syndrome, Guangzhou, PR China.
| |
Collapse
|
2
|
Kan S, Feng S, Zhao X, Chen Z, Zhou M, Liu L, Zhu H, Cheng Y, Fu X, Hu W, Zhu R. UAMC-3203 inhibits ferroptosis and promotes functional recovery in rats with spinal cord injury. Sci Rep 2024; 14:20180. [PMID: 39215144 PMCID: PMC11364804 DOI: 10.1038/s41598-024-70926-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Spinal cord injury (SCI) results in irreversible neurological impairment. After SCI, Ferritinophagy-induced free iron released from ferritin can lead to extensive lipid peroxidation and aggravate neurological damage. NRF2/HO-1 pathway is to endow cells with a protective effect against oxidative stress, and it plays an important role in the transcriptional activation of a series of antioxidant and detoxification genes. UAMC-3203 is a ferrostatin-1(Fer-1) analogue with better solubility and stability, which can more effectively inhibit ferroptosis after SCI. A rat SCI model was constructed, and the recovery of motor function was observed after treatment with UAMC-3203. ELISA was employed to assess the impact of UAMC-3203 on inflammation-related factors, while immunofluorescence was utilized to investigate the influence of UAMC-3203 on neuronal count as well as the activation of astrocytes and microglia/macrophages. Malondialdehyde (MDA) were detected to reflect the level of oxidation products. Western blot analysis was used to measure the level of ferroptosis markers and the expression of NRF2/HO-1. Our findings demonstrate that UAMC-3203 inhibits the production of reactive oxygen species (ROS) and lipid peroxides, preventing ferroptosis and reducing neuronal degeneration. Additionally, UAMC-3203 suppresses astrocyte proliferation and microglia/macrophage activation, as well as the release of ferroptosis-related inflammatory factors. These combined effects contribute to the preservation of spinal cord tissue and the facilitation of motor function recovery. UAMC-3203 maybe inhibit ferroptosis after SCI to promote functional recovery.
Collapse
Affiliation(s)
- Shunli Kan
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin, China
| | - Sa Feng
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin, China
| | - Xinyan Zhao
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin, China
| | - Ziyu Chen
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin, China
| | - Mengmeng Zhou
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin, China
| | - Linyan Liu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin, China
| | - Haoqiang Zhu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin, China
| | - Yuelin Cheng
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin, China
| | - Xuanhao Fu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin, China
| | - Wei Hu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin, China
| | - Rusen Zhu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China.
- Tianjin Institute of Spinal Surgery, Tianjin, China.
| |
Collapse
|
3
|
Li X, Jiang Y, Wang Y, Li N, Zhang S, Lv K, Jia R, Wei T, Li X, Han C, Lin J. KLF4 suppresses anticancer effects of brusatol via transcriptional upregulating NCK2 expression in melanoma. Biochem Pharmacol 2024; 223:116197. [PMID: 38583810 DOI: 10.1016/j.bcp.2024.116197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/21/2024] [Accepted: 04/05/2024] [Indexed: 04/09/2024]
Abstract
Brusatol (Bru), a main extract from traditional Chinese medicine Brucea javanica, has been reported to exist antitumor effect in many tumors including melanoma. However, the underlying mechanism in its anti-melanoma effect still need further exploration. Here, we reported that the protein expression of KLF4 in melanoma cells were significantly downregulated in response to brusatol treatment. Overexpression of KLF4 suppressed brusatol-induced melanoma cell apoptosis; while knockdown of KLF4 enhanced antitumor effects of brusatol on melanoma cells not only in vitro but also in vivo. Further studies on the mechanism revealed that KLF4 bound to the promoter of NCK2 directly and facilitated NCK2 transcription, which suppressed the antitumor effect of brusatol on melanoma. Furthermore, our findings showed that miR-150-3p was dramatically upregulated under brusatol treatment which resulted in the downregulation of KLF4. Our results suggested that the miR-150-3p/KLF4/NCK2 axis might play an important role in the antitumour effects of brusatol in melanoma.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian 116044, PR China; Institute of Cancer Stem Cell of Dalian Medical University, Dalian 116044, PR China
| | - Yuankuan Jiang
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian 116044, PR China
| | - Ying Wang
- Institute of Cancer Stem Cell of Dalian Medical University, Dalian 116044, PR China
| | - Na Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian 116044, PR China
| | - Shumeng Zhang
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian 116044, PR China
| | - Kejia Lv
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian 116044, PR China
| | - Renchuan Jia
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian 116044, PR China
| | - Tianfu Wei
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian 116044, PR China
| | - Xiaojie Li
- College of Stomatology Dalian Medical University, Dalian 116044, PR China.
| | - Chuanchun Han
- Institute of Cancer Stem Cell of Dalian Medical University, Dalian 116044, PR China.
| | - Jingrong Lin
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian 116044, PR China.
| |
Collapse
|
4
|
Yang Z, Zhong H, Wu Y, Li S, Gong S, Cai Y, Jin J. Design and Preparation of Iodinated Brucea Javanica Oil as Dual Functional Anti-Tumor Agent for Treating Hepatocellular Carcinoma. Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02717-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
5
|
Dai Z, Cai L, Chen Y, Wang S, Zhang Q, Wang C, Tu M, Zhu Z, Li Q, Lu X. Brusatol Inhibits Proliferation and Invasion of Glioblastoma by Down-Regulating the Expression of ECM1. Front Pharmacol 2022; 12:775680. [PMID: 34970146 PMCID: PMC8713816 DOI: 10.3389/fphar.2021.775680] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/30/2021] [Indexed: 12/13/2022] Open
Abstract
Brusatol (Bru), a Chinese herbal extract, has a variety of anti-tumor effects. However, little is known regarding its role and underlying mechanism in glioblastoma cells. Here, we found that Bru could inhibit the proliferation of glioblastoma cells in vivo and in vitro. Besides, it also had an inhibitory effect on human primary glioblastoma cells. RNA-seq analysis indicated that Bru possibly achieved these effects through inhibiting the expression of extracellular matrix protein 1 (ECM1). Down-regulating the expression of ECM1 via transfecting siRNA could weaken the proliferation and invasion of glioblastoma cells and promote the inhibitory effect of Bru treatment. Lentivirus-mediated overexpression of ECM1 could effectively reverse this weakening effect. Our findings indicated that Bru could inhibit the proliferation and invasion of glioblastoma cells by suppressing the expression of ECM1, and Bru might be a novel effective anticancer drug for glioblastoma cells.
Collapse
Affiliation(s)
- Zhang'an Dai
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lin Cai
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yingyu Chen
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Silu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengde Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ming Tu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhangzhang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Li
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xianghe Lu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Cheng C, Yuan F, Chen XP, Zhang W, Zhao XL, Jiang ZP, Zhou HH, Zhou G, Cao S. Inhibition of Nrf2-mediated glucose metabolism by brusatol synergistically sensitizes acute myeloid leukemia to Ara-C. Biomed Pharmacother 2021; 142:111652. [PMID: 34112534 DOI: 10.1016/j.biopha.2021.111652] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/13/2021] [Accepted: 04/21/2021] [Indexed: 12/20/2022] Open
Abstract
Chemotherapy resistance remains to be the primary barrier to acute myeloid leukemia (AML) treatment failure. Nuclear factor-erythroid 2-related factor 2 (Nrf2) has been well established as a truly pleiotropic transcription factor. Inhibition of Nrf2 function increases the sensitivity of various chemotherapeutics and overcomes chemoresistance effectively. Brusatol (Bru) has been reported to decrease Nrf2 protein expression specifically by ubiquitin degradation of Nrf2. However, it remains elusive whether combination of Brusatol and Cytarabine (Ara-C) elicits a synergistic antitumor effect in AML. Our results demonstrated that combination of Ara-C and Brusatol synergistically exerted remarkable pro-apoptosis effect in HL-60 and THP-1 cells. Mechanistically, synergistic anti-tumor effect of Ara-C/Brusatol in AML cells is mediated by attenuating Nrf2 expression. To our surprise, Nrf2 inhibition by Brusatol causes downregulation of the expression of glycolysis-related proteins and decreased glucose consumption and lactate production, whereas the level of ROS production was unaffected. The activation of Nrf2 by Sulforaphane (SFP) could reverse the chemotherapeutic effect and changes of glycolysis of concomitant of Ara-C with Brusatol in AML cell lines. Additionally, Ara-C/Brusatol co-treatment decreased Glucose-6-phosphate dehydrogenase (G6PD) protein expression and increased the sensitivity of Ara-C. Moreover, the mouse xenograft in vivo experiment confirmed that combining Ara-C with Brusatol exerted stronger antileukemia than Ara-C alone. The efficacy, together with the mechanistic observations, reveals the potential of simultaneously giving these two drugs and provides a rational basis for targeting glucose catabolism in future clinical therapeutic approach.
Collapse
Affiliation(s)
- Cong Cheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Center South University, Changsha 410008, Hunan, PR China
| | - Fang Yuan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Center South University, Changsha 410008, Hunan, PR China
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Center South University, Changsha 410008, Hunan, PR China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Center South University, Changsha 410008, Hunan, PR China
| | - Xie-Lan Zhao
- Department of Hematology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China
| | - Zhi-Ping Jiang
- Department of Hematology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Center South University, Changsha 410008, Hunan, PR China
| | - Gan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Center South University, Changsha 410008, Hunan, PR China; National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, 110 Xiangya Road, Changsha, Hunan 410008, PR China.
| | - Shan Cao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Center South University, Changsha 410008, Hunan, PR China.
| |
Collapse
|
7
|
Wang H, Tu WJ, Xiao C, Dong MX, Ye YT, Deng J, Wang Y, Sha H, Liu Q. Nrf2 played an important role in radiation protection effect of low-level laser exposed on umbilical cord mesenchymal stem cell. Tissue Cell 2020; 63:101329. [PMID: 32223956 DOI: 10.1016/j.tice.2019.101329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/31/2019] [Accepted: 12/31/2019] [Indexed: 11/19/2022]
Abstract
To investigate the protective function of low-level laser irradiation (LLLI) against ionizing irradiation and explore the molecular mechanism of photomodulation of Nrf2 protein, the impact of LLLI (635 nm, 5.7 J/cm2) before 2 Gy gamma ray radiation of radio-sensitive tissue hematopoietic stem cells was evaluated. As a result, reduced levels of reactive oxygen species and increased expression of antioxidant enzymes were detected. Moreover, increased expression of Nrf2 was observed after LLLI, whereas brusatol pretreatment before LLLI abolished this effect. In vivo, transplantation of human umbilical cord mesenchymal stem cells (hUC-MSCs) was employed for therapy of hematopoietic function in an acute radiation sickness (H-ARS) mouse model, which was induced by 6-Gy ionizing irradiation; different hUC-MSC pretreatments including LLLI and Nrf2 RNAi were accounted for during experimental grouping. LLLI treatment of cells significantly increased the erythrocyte count and number of myelopoiesis clones (P < 0.05), but such improvements were reduced by Nrf2 RNAi pretreatment compared with cells transplanted without intervention. Therefore, LLLI may improve the radiation protection effect through molecular mechanisms related to the Nrf2 antioxidant pathway.
Collapse
Affiliation(s)
- Hong Wang
- Geriatric Health Engineering Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Wen-Jun Tu
- Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Changyan Xiao
- Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Ming-Xin Dong
- Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Yin-Tao Ye
- Cancer Institute & Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Juan Deng
- Geriatric Health Engineering Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Yan Wang
- Geriatric Health Engineering Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Hong Sha
- Geriatric Health Engineering Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China.
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China.
| |
Collapse
|
8
|
Lee CH. Reversal of Epithelial-Mesenchymal Transition by Natural Anti-Inflammatory and Pro-Resolving Lipids. Cancers (Basel) 2019; 11:E1841. [PMID: 31766574 PMCID: PMC6966475 DOI: 10.3390/cancers11121841] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 02/08/2023] Open
Abstract
Epithelial mesenchymal transition (EMT) is a key process in the progression of malignant cancer. Therefore, blocking the EMT can be a critical fast track for the development of anticancer drugs. In this paper, we update recent research output of EMT and we explore suppression of EMT by natural anti-inflammatory compounds and pro-resolving lipids.
Collapse
Affiliation(s)
- Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 100-715, Korea
| |
Collapse
|
9
|
Turpaev K, Krizhanovskii C, Wang X, Sargsyan E, Bergsten P, Welsh N. The protein synthesis inhibitor brusatol normalizes high-fat diet-induced glucose intolerance in male C57BL/6 mice: role of translation factor eIF5A hypusination. FASEB J 2019; 33:3510-3522. [PMID: 30462531 DOI: 10.1096/fj.201801698r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The naturally occurring quassinoid compound brusatol improves the survival of insulin-producing cells when exposed to the proinflammatory cytokines IL-1β and IFN-γ in vitro. The aim of the present study was to investigate whether brusatol also promotes beneficial effects in mice fed a high-fat diet (HFD), and if so, to study the mechanisms by which brusatol acts. In vivo, we observed that the impaired glucose tolerance of HFD-fed male C57BL/6 mice was counteracted by a 2 wk treatment with brusatol. Brusatol treatment improved both β-cell function and peripheral insulin sensitivity of HFD-fed mice. In vitro, brusatol inhibited β-cell total protein and proinsulin biosynthesis, with an ED50 of ∼40 nM. In line with this, brusatol blocked cytokine-induced iNOS protein expression via inhibition of iNOS mRNA translation. Brusatol may have affected protein synthesis, at least in part, via inhibition of eukaryotic initiation factor 5A (eIF5A) hypusination, as eIF5A spermidine association and hypusination in RIN-5AH cells was reduced in a dose- and time-dependent manner. The eIF5A hypusination inhibitor GC7 promoted a similar effect. Both brusatol and GC7 protected rat RIN-5AH cells against cytokine-induced cell death. Brusatol reduced eIF5A hypusination and cytokine-induced cell death in EndoC-βH1 cells as well. Finally, hypusinated eIF5A was reduced in vivo by brusatol in islet endocrine and endothelial islet cells of mice fed an HFD. The results of the present study suggest that brusatol improves glucose intolerance in mice fed an HFD, possibly by inhibiting protein biosynthesis and eIF5A hypusination.-Turpaev, K., Krizhanovskii, C., Wang, X., Sargsyan, E., Bergsten, P., Welsh, N. The protein synthesis inhibitor brusatol normalizes high-fat diet-induced glucose intolerance in male C57BL/6 mice: role of translation factor eIF5A hypusination.
Collapse
Affiliation(s)
- Kyril Turpaev
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Camilla Krizhanovskii
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Xuan Wang
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Ernest Sargsyan
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Peter Bergsten
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Nils Welsh
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| |
Collapse
|
10
|
Cao SY, Ye SJ, Wang WW, Wang B, Zhang T, Pu YQ. Progress in active compounds effective on ulcerative colitis from Chinese medicines. Chin J Nat Med 2019; 17:81-102. [PMID: 30797423 DOI: 10.1016/s1875-5364(19)30012-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Indexed: 02/09/2023]
Abstract
Ulcerative colitis (UC), a chronic inflammatory disease affecting the colon, has a rising incidence worldwide. The known pathogenesis is multifactorial and involves genetic predisposition, epithelial barrier defects, dysregulated immune responses, and environmental factors. Nowadays, the drugs for UC include 5-aminosalicylic acid, steroids, and immunosuppressants. Long-term use of these drugs, however, may cause several side effects, such as hepatic and renal toxicity, drug resistance and allergic reactions. Moreover, the use of traditional Chinese medicine (TCM) in the treatment of UC shows significantly positive effects, low recurrence rate, few side effects and other obvious advantages. This paper summarizes several kinds of active compounds used in the experimental research of anti-UC effects extracted from TCM, mainly including flavonoids, acids, terpenoids, phenols, alkaloids, quinones, and bile acids from some animal medicines. It is found that the anti-UC activities are mainly focused on targeting inflammation or oxidative stress, which is associated with increasing the levels of anti-inflammatory cytokine (IL-4, IL-10, SOD), suppressing the levels of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-8, IL-23, NF-κB, NO), reducing the activity of MPO, MDA, IFN-γ, and iNOS. This review may offer valuable reference for UC-related studies on the compounds from natural medicines.
Collapse
Affiliation(s)
- Si-Yu Cao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Sheng-Jie Ye
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei-Wei Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-Qiong Pu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
11
|
Mabhida SE, Dludla PV, Johnson R, Ndlovu M, Louw J, Opoku AR, Mosa RA. Protective effect of triterpenes against diabetes-induced β-cell damage: An overview of in vitro and in vivo studies. Pharmacol Res 2018; 137:179-192. [PMID: 30315968 DOI: 10.1016/j.phrs.2018.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/27/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022]
Abstract
Accumulative evidence shows that chronic hyperglycaemia is a major factor implicated in the development of pancreatic β-cell dysfunction in diabetic patients. Furthermore, most of these patients display impaired insulin signalling that is responsible for accelerated pancreatic β-cell damage. Indeed, prominent pathways involved in glucose metabolism such as phosphatidylinositol 3-kinase/ protein kinase B (PI3-K/AKT) and 5' AMP-activated protein kinase (AMPK) are impaired in an insulin resistant state. The impairment of this pathway is associated with over production of reactive oxygen species and pro-inflammatory factors that supersede pancreatic β-cell damage. Although several antidiabetic drugs can improve β-cell function by modulating key regulators such as PI3-K/AKT and AMPK, evidence of their β-cell regenerative and protective effect is scanty. As a result, there has been continued exploration of novel antidiabetic therapeutics with abundant antioxidant and antiinflammatory properties that are essential in protecting against β-cell damage. Such therapies include triterpenes, which have displayed robust effects to improve glycaemic tolerance, insulin secretion, and pancreatic β-cell function. This review summarises most relevant effects of various triterpenes on improving pancreatic β-cell function in both in vitro and in vivo experimental models. A special focus falls on studies reporting on the ameliorative properties of these compounds against insulin resistance, oxidative stress and inflammation, the well-known factors involved in hyperglycaemia associated tissue damage.
Collapse
Affiliation(s)
- Sihle E Mabhida
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, 3886, South Africa; Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, 7505, South Africa.
| | - Phiwayinkosi V Dludla
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, 60121, Italy; Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, 7505, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, 7505, South Africa; Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, Stellenbosch, South Africa
| | - Musawenkosi Ndlovu
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, 3886, South Africa
| | - Johan Louw
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, 3886, South Africa; Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, 7505, South Africa
| | - Andy R Opoku
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, 3886, South Africa
| | - Rebamang A Mosa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, 3886, South Africa
| |
Collapse
|
12
|
Guo N, Xu X, Yuan G, Chen X, Wen Q, Guo R. Pharmacokinetic, metabolic profiling and elimination of brusatol in rats. Biomed Chromatogr 2018; 32:e4358. [PMID: 30089336 DOI: 10.1002/bmc.4358] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/25/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022]
Abstract
Brusatol, a quassinoid isolated from the traditional Chinese medicine Brucea javanica, has been reported to be an inhibitor of Nrf2 pathway and has great potential to be developed into a novel chemotherapeutic adjuvant. However, the in vivo process of brusatol has not been comprehensively explained yet. Therefore, this paper focused on the pharmacokinetic metabolism and excretion of brusatol in rats using a simple and reproducible LC-MS/MS method. The results indicated that the plasma concentration of brusatol decreased rapidly; the average cumulative excretion rate in urine was 5.82% during 24 h, and 0.71% in bile during 12 h. High-resolution mass spectrometry was applied for the identification of metabolites; as a result, four metabolites were detected and the structure was tentatively deduced on the base of the MS2 data, Compound Discoverer 2.0 and Mass Frontier 7.0 software. Hydroxylation, hydrolysis and glucuronidation were suggested as major metabolic pathways in vivo. The in vivo process and detection of metabolites of brusatol might improve the understanding of the mechanism of its anticancer effect and provide valuable information for its safety estimation, which will be essential to the new drug development.
Collapse
Affiliation(s)
- Nan Guo
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Xu
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Guiyan Yuan
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, China
| | - Xuwang Chen
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, China
| | - Qing Wen
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Ruichen Guo
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
13
|
Man F, Choo CY. Safety assessment of standardized aqueous Brucea javanica extract in rats. JOURNAL OF ETHNOPHARMACOLOGY 2018; 215:21-26. [PMID: 29288829 DOI: 10.1016/j.jep.2017.12.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/06/2017] [Accepted: 12/26/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The seeds of Brucea javanica and its aqueous decoction is a traditional medicine consumed by diabetic patients in Malaysia. The daily consumption of B. javanica seeds and it's aqueous decoction causes much concern as the quassinoids and its glycosides from the seeds exhibited various pharmacological activity at low doses. AIMS OF STUDY The aim of the present study is to evaluate the repeated dose toxicity of the standardized aqueous extract administered daily for 30 days through oral administration at its effective hypoglycemia doses. MATERIALS AND METHODS The seeds were dried, ground and extracted in deionized water. A HPLC-photodiode array method was developed and validated for the standardization of both the hypoglycemia agents, namely bruceine D and E in aqueous extract. Both normoglycemia and streptozotocin (STZ)-induced diabetic rats were fed orally with 15, 30 and 60mg/kg body weight of standardized aqueous extract. The blood glucose was measured at 0-8h. In repeated dose toxicity, similar doses were administered orally to rats for 30 days. At the end of 30 days, the blood was withdrawn and subjected to biochemical and haematology analysis while organs were harvested for histology analysis. RESULTS Oral administration of standardized aqueous extract exhibited a dose-response relationship in both the normoglycemia and STZ-induced diabetic rats. Daily oral administration of 15, 30 and 60mg/kg standardized aqueous extract for 30 days to rats did not show signs to toxicity in its biochemical, haematology and histology analysis. CONCLUSION In conclusion, although the seeds were reported to contain compounds with various pharmacological activity, the daily oral administration to rats for 30 days do not showed signs of toxicity at its effective hypoglycemia doses.
Collapse
Affiliation(s)
- Farahdina Man
- MedChem Herbal Research Group, Faculty of Pharmacy, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300 Puncak Alam, Selangor, Malaysia
| | - Chee-Yan Choo
- MedChem Herbal Research Group, Faculty of Pharmacy, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300 Puncak Alam, Selangor, Malaysia.
| |
Collapse
|
14
|
Zhou J, Tan L, Xie J, Lai Z, Huang Y, Qu C, Luo D, Lin Z, Huang P, Su Z, Xie Y. Characterization of brusatol self-microemulsifying drug delivery system and its therapeutic effect against dextran sodium sulfate-induced ulcerative colitis in mice. Drug Deliv 2018; 24:1667-1679. [PMID: 29078713 PMCID: PMC8253134 DOI: 10.1080/10717544.2017.1384521] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Brusatol (BR) is one of the main bioactive components derived from Brucea javanica, a medicinal herb historically used in the treatment of dysenteric disorders (also known as ulcerative colitis(UC)). Due to its poor aqueous solubility, a novel brusatol self-microemulsifying drug delivery system (BR-SMEDDS) nanoformulation with smaller size, higher negative zeta potential and drug content, and excellent stability was developed. The appearance of BR-SMEDDS remained clear and transparent, and transmission electron microscopy showed microemulsion droplets to be spherical with homogeneous distribution. Pharmacokinetic parameters indicated that oral bioavailability was greatly improved by BR-SMEDDS as compared with aqueous suspension. Meanwhile, the anti-colitis activity of BR-SMEDDS was evaluated on dextran sodium sulfate (DSS)-induced colitis mice model. The result illustrated that the nano-formation significantly reduced the body weight loss, recovered colon length, decreased disease activity index and microscopic score, regulated immune-inflammatory cytokines, diminished oxidative stress and repressed the colonic expression of myeloid differentiation factor 88 (MyD88), toll-like receptor 4 (TLR4) and nuclear factor kappa B p65 (NF-κB p65) proteins. Our findings demonstrated for the first time that BR could effectively attenuate colonic inflammation in mice, at least partially, via favorable regulation of anti-oxidative and anti-inflammatory status and inhibition of the TLR4-linked NF-κB signaling pathway. The BR nano-formulation was superior to BR suspension and sulphasalazine, in treating experimental UC, and exhibited similar effect with azathioprine, with much smaller dosage. The enhanced anti-UC effect of BR might be intimately associated with the improved pharmacokinetic property by SMEDDS. The developed nano-delivery system might thus be a promising candidate for colitis treatment.
Collapse
Affiliation(s)
- Jiangtao Zhou
- a School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , PR China.,b Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , PR China
| | - Lihua Tan
- b Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , PR China
| | - Jianhui Xie
- c Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome , The Second Affiliated Hospital, Guangzhou University of Chinese Medicine , Guangzhou , PR China
| | - Zhengquan Lai
- d School of Chinese Medicine, Faculty of Medicine , The Chinese University of Hong Kong , Hong Kong , PR China
| | - Yanfeng Huang
- b Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , PR China
| | - Chang Qu
- b Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , PR China
| | - Dandan Luo
- b Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , PR China
| | - Zhixiu Lin
- d School of Chinese Medicine, Faculty of Medicine , The Chinese University of Hong Kong , Hong Kong , PR China
| | - Ping Huang
- a School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , PR China
| | - Ziren Su
- b Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , PR China
| | - Youliang Xie
- b Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , PR China
| |
Collapse
|
15
|
Chen HM, Lai ZQ, Liao HJ, Xie JH, Xian YF, Chen YL, Ip SP, Lin ZX, Su ZR. Synergistic antitumor effect of brusatol combined with cisplatin on colorectal cancer cells. Int J Mol Med 2018; 41:1447-1454. [PMID: 29328398 PMCID: PMC5819912 DOI: 10.3892/ijmm.2018.3372] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 12/21/2017] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is a common and life-threatening type of malignant cancer, which is associated with a high mortality rate. Cisplatin (CDDP) is a commonly used chemotherapy drug with significant side effects. Brusatol (BR) is one of the principal chemical compounds isolated from the Chinese herb Bruceae Fructus, which has been reported to markedly inhibit the proliferation of numerous cancer cell lines. The present study aimed to investigate the possible synergistic anticancer effects of CDDP combined with BR on CT-26 cells, and to evaluate the underlying mechanisms of action. The growth inhibitory effects of BR, CDDP, and BR and CDDP cotreatment on CT-26 cells were assessed by MTT assay. Cell apoptosis were determined by flow cytometry and western blot analysis. The results indicated that compared with single-agent treatment, cotreatment of CT-26 cells with CDDP and BR synergistically inhibited cell proliferation and increased cellular apoptosis. Furthermore, treatment of CT-26 cells with CDDP and BR resulted in a marked increase in the release of cytosolic cytochrome c, decreased expression of procaspase-3 and procaspase-9, and upregulation of the B-cell lymphoma 2 (Bcl-2)-associated X protein/Bcl-2 ratio compared with treatment with BR or CDDP alone. These results strongly suggested that the combination of CDDP and BR was able to produce a synergistic antitumor effect in CRC cells, thus providing a solid foundation for further development of this combination regimen into an effective therapeutic method for CRC.
Collapse
Affiliation(s)
- Hai-Ming Chen
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Zheng-Quan Lai
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR 999077, P.R. China
| | - Hui-Jun Liao
- Department of Clinical Pharmacy and Pharmaceutical Services, Shenzhen Sixth People's Hospital (Nanshan Hospital), Shenzhen 518060, P.R. China
| | - Jian-Hui Xie
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR 999077, P.R. China
| | - Yun-Long Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR 999077, P.R. China
| | - Siu-Po Ip
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR 999077, P.R. China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR 999077, P.R. China
| | - Zi-Ren Su
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
16
|
Nicco C, Batteux F. ROS Modulator Molecules with Therapeutic Potential in Cancers Treatments. Molecules 2017; 23:E84. [PMID: 29301225 PMCID: PMC6016996 DOI: 10.3390/molecules23010084] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 12/28/2017] [Accepted: 12/30/2017] [Indexed: 12/11/2022] Open
Abstract
Reactive Oxygen Species (ROS) are chemically reactive chemical species containing oxygen. The redox status of a cell is function of the relative concentrations of oxidized and reduced forms of proteins, enzymes, ROS, molecules containing thiol and other factors. In the organism, the redox balance is based on the generation and elimination of ROS produced by endogenous and exogenous sources. All living organisms must maintain their redox equilibrium to survive and proliferate. Enzymatic and molecular pathways control ROS levels tightly but differentially depending on the type of cell. This review is an overview of various molecules that modulate ROS production/detoxification and have a synergistic action with the chemotherapies to kill cancer cells while preserving normal cells to avoid anticancer drugs side effects, allowing a better therapeutic index of the anticancer treatments.
Collapse
Affiliation(s)
- Carole Nicco
- Department "Development, Reproduction and Cancer", Cochin Institute, INSERM U1016, University Paris Descartes, Paris 75014, France.
| | - Frédéric Batteux
- Department "Development, Reproduction and Cancer", Cochin Institute, INSERM U1016, University Paris Descartes, Paris 75014, France.
| |
Collapse
|
17
|
Malainer C, Schachner D, Sangiovanni E, Atanasov AG, Schwaiger S, Stuppner H, Heiss EH, Dirsch VM. Eurycomalactone Inhibits Expression of Endothelial Adhesion Molecules at a Post-Transcriptional Level. JOURNAL OF NATURAL PRODUCTS 2017; 80:3186-3193. [PMID: 29148754 PMCID: PMC5744186 DOI: 10.1021/acs.jnatprod.7b00503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The C-19 quassinoid eurycomalactone (1) has recently been shown to be a potent (IC50 = 0.5 μM) NF-κB inhibitor in a luciferase reporter model. In this study, we show that 1 with similar potency inhibited the expression of the NF-κB-dependent target genes ICAM-1, VCAM-1, and E-selectin in TNFα-activated human endothelial cells (HUVECtert) by flow cytometry experiments. Surprisingly, 1 (2 μM) did not inhibit TNFα-induced IKKα/β or IκBα phosphorylation significantly. Also, the TNFα-induced degradation of IκBα remained unchanged in response to 1 (2 μM). In addition, pretreatment of HUVECtert with 1 (2 μM) had no statistically significant effect on TNFα-mediated nuclear translocation of the NF-κB subunit p65 (RelA). Quantitative RT-PCR revealed that 1 (0.5-5 μM) exhibited diverse effects on the TNFα-induced transcription of ICAM-1, VCAM-1, and SELE genes since the mRNA level either remained unchanged (ICAM-1, E-selectin, and VCAM-1 at 0.5 μM 1), was reduced (VCAM-1 at 5 μM 1), or even increased (E-selectin at 5 μM 1). Finally, the time-dependent depletion of a short-lived protein (cyclin D1) as well as the measurement of de novo protein synthesis in the presence of 1 (2-5 μM) suggested that 1 might act as a protein synthesis inhibitor rather than an inhibitor of early NF-κB signaling.
Collapse
Affiliation(s)
- Clemens Malainer
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Daniel Schachner
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Enrico Sangiovanni
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
- Department
of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy
| | - Atanas G. Atanasov
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
- Institute
of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552 Jastrzebiec, Poland
| | - Stefan Schwaiger
- Institute
of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck, University Innsbruck, Innrain 80/82, Innsbruck 6020, Austria
| | - Hermann Stuppner
- Institute
of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck, University Innsbruck, Innrain 80/82, Innsbruck 6020, Austria
| | - Elke H. Heiss
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Verena M. Dirsch
- Department
of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
- Tel: +43-1-4277-55270. Fax: +43-1-4277-55969. E-mail:
| |
Collapse
|
18
|
Oh ET, Kim CW, Kim HG, Lee JS, Park HJ. Brusatol-Mediated Inhibition of c-Myc Increases HIF-1α Degradation and Causes Cell Death in Colorectal Cancer under Hypoxia. Am J Cancer Res 2017; 7:3415-3431. [PMID: 28912885 PMCID: PMC5596433 DOI: 10.7150/thno.20861] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/20/2017] [Indexed: 01/09/2023] Open
Abstract
HIF-1 (hypoxia-inducible factor-1) regulates the expression of ~100 genes involved in angiogenesis, metastasis, tumor growth, chemoresistance and radioresistance, underscoring the growing interest in targeting HIF-1 for cancer control. In the present study, we investigated the molecular mechanisms underlying brusatol-induced HIF-1α degradation and cell death in colorectal cancer under hypoxia (0.5% O2). Under hypoxia, pretreatment of cancer cells with brusatol increased HIF-1α degradation and cancer cell death in a dose-dependent manner. This effect was mediated by activation of prolyl hydroxylases (PHDs), as evidenced by the block of brusatol-induced HIF-1α degradation and cancer cell death by both pharmacological inhibition and siRNA-mediated knockdown of PHDs. In addition, a ferrous iron chelator (2,2'-bypyridyl) blocked brusatol-induced degradation of HIF-1α and cancer cell death in hypoxia by inhibiting PHD activation. We further found that brusatol inhibited c-Myc expression, and showed that overexpression of c-Myc prevented brusatol-induced degradation of HIF-1α and cancer cell death by increasing mitochondrial ROS production and subsequent ROS-mediated transition of ferrous iron to ferric iron. Consistent with these results, treatment of tumor-bearing mice with brusatol significantly suppressed tumor growth by promoting PHD-mediated HIF-1α degradation. Collectively, our results suggest that brusatol-mediated inhibition of c-Myc/ROS signaling pathway increases HIF-1α degradation by promoting PHD activity and induces cell death in colorectal cancer under hypoxia
Collapse
|
19
|
Krizhanovskii C, Kristinsson H, Elksnis A, Wang X, Gavali H, Bergsten P, Scharfmann R, Welsh N. EndoC-βH1 cells display increased sensitivity to sodium palmitate when cultured in DMEM/F12 medium. Islets 2017; 9:e1296995. [PMID: 28277987 PMCID: PMC5465947 DOI: 10.1080/19382014.2017.1296995] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Aims - Human pancreatic islets are known to die in response to the free fatty acid of sodium palmitate when cultured in vitro. This is in contrast to EndoC-βH1 cells, which in our hands are not sensitive to the cell death-inducing effects sodium palmitate, making these cells seemingly unsuitable for lipotoxicity studies. However, the EndoC-βH1 cells are routinely cultured in a nutrient mixture based on Dulbecco's Modified Eagle Medium (DMEM), which may not be the optimal choice for studies dealing with lipotoxicity. The aim of the present investigation was to define culture conditions that render EndoC-βH1 cells sensitive to toxic effects of sodium palmitate. Methods - EndoC-βH1 cells were cultured at standard conditions in either DMEM or DMEM/F12 culture medium. Cell death was analyzed using propidium iodide staining and flow cytometry. Insulin release and content was quantified using a human insulin ELISA. Results - We presently observe that substitution of DMEM for a DMEM/Ham's F12 mixture (50%/50% vol/vol) renders the cells sensitive to the apoptotic effects of sodium palmitate and sodium palmitate + high glucose leading to an increased cell death. Supplementation of the DMEM culture medium with linoleic acid partially mimicked the effect of DMEM/F12. Culture of EndoC-βH1 cells in DMEM/F12 resulted also in increased proliferation, ROS production and insulin contents, but markers for metabolic stress, autophagy or amyloid deposits were unaffected. Conclusions - The culture conditions for EndoC-βH1 cells can be modified so these cells display signs of lipotoxicity in response to sodium palmitate.
Collapse
Affiliation(s)
- Camilla Krizhanovskii
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Hjalti Kristinsson
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Andris Elksnis
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Xuan Wang
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Hamid Gavali
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Peter Bergsten
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Raphael Scharfmann
- INSERM, U1016, Institut Cochin, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nils Welsh
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- CONTACT Nils Welsh , Science for Life Laboratory, Department of Medical Cell Biology, Box 571, BMC, SE-751 23 Uppsala, Sweden
| |
Collapse
|
20
|
Xiang Y, Ye W, Huang C, Lou B, Zhang J, Yu D, Huang X, Chen B, Zhou M. Brusatol inhibits growth and induces apoptosis in pancreatic cancer cells via JNK/p38 MAPK/NF-κb/Stat3/Bcl-2 signaling pathway. Biochem Biophys Res Commun 2017; 487:820-826. [PMID: 28455228 DOI: 10.1016/j.bbrc.2017.04.133] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 12/18/2022]
Abstract
Brusatol, isolated from brucea, has been proved to exhibit anticancer influence on various kind of human malignancies. However, the role that brusatol plays in pancreatic cancer is seldom known by the public. Through researches brusatol was proved to inhibit growth and induce apoptosis in both PATU-8988 and PANC-1 cells by decreasing the expression level of Bcl-2 and increasing the expression levels of Bax, Cleaved Caspase-3. Then we found the activation of the JNK, p38 MAPK and inactivation of the NF-κb, Stat3 are related with the potential pro-apoptotic signaling pathways. However, SP600125 could not only abrogated the JNK activation caused by brusatol, but also reverse the p38 activation and the decrease of Bcl-2 as SB203580 did. Besides, SP600125 and SB203580 also reversed the inactivation of NF-κb and Stat3. Furthermore, BAY 11-7082 and S3I-201 indeed had the similar effect as brusatol had on the expression of Phospho-Stat3 and Bcl-2. To sum up, we came to a conclusion that in pancreatic cancer, brusatol do inhibit growth and induce apoptosis. And we inferred that brusatol illustrates anticancer attribution via JNK/p38 MAPK/NF-κb/Stat3/Bcl-2 signaling pathway.
Collapse
Affiliation(s)
- Yukai Xiang
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Wen Ye
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Chaohao Huang
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Bin Lou
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Jie Zhang
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Dinglai Yu
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Xince Huang
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Bicheng Chen
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China; Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Wenzhou, Zhejiang Province, People's Republic of China
| | - Mengtao Zhou
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China.
| |
Collapse
|
21
|
Scharfmann R, Didiesheim M, Richards P, Chandra V, Oshima M, Albagli O. Mass production of functional human pancreatic β-cells: why and how? Diabetes Obes Metab 2016; 18 Suppl 1:128-36. [PMID: 27615142 DOI: 10.1111/dom.12728] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022]
Abstract
Diabetes (either type 1 or type 2) is due to insufficient functional β-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either β-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model β-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human β-cells share many similarities with rodent β-cells, they also differ on a number of important parameters. In this context, developing ways to study human β-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human β-cells that should be useful tools to discover new ways to modulate functional β-cell mass in humans.
Collapse
Affiliation(s)
- R Scharfmann
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France.
| | - M Didiesheim
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - P Richards
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - V Chandra
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - M Oshima
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - O Albagli
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| |
Collapse
|
22
|
Zhang Q, Yuan Y, Cui J, Xiao T, Deng Z, Jiang D. Determination of a potential antitumor quassinoid in rat plasma by UPLC-MS/MS and its application in a pharmacokinetic study. J Pharm Biomed Anal 2016; 124:143-148. [PMID: 26945636 DOI: 10.1016/j.jpba.2016.02.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/25/2016] [Accepted: 02/28/2016] [Indexed: 10/22/2022]
Abstract
A sensitive UPLC-MS/MS method was developed and validated for the determination of brusatol in rat plasma. Chromatographic separation was carried out on a C18 column using methanol and 10mM ammonium acetate containing 0.1% (v/v) formic acid (55:45, v/v). The lower limit of quantification (LLOQ) was 1.0 ng/mL for brusatol in plasma. The intra- and inter-day precision for the analyte ranged from 3.2% to 9.2% and 1.3% to 7.8%, and the accuracy was between 97.3% and 108.5%. The method was successfully applied in a pharmacokinetic study of brusatol following intravenous injection (0.5, 1.0, and 2.0mg/kg) of brusatol.
Collapse
Affiliation(s)
- Qiang Zhang
- Breast Surgery, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Yonghui Yuan
- Department of Infection, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Jianchun Cui
- Department of Endocrine Surgery, the People's Hospital of Liaoning Province, Shenyang 110016, China
| | - Tingting Xiao
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Zhipeng Deng
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan 250062, China.
| | - Daqing Jiang
- Breast Surgery, Liaoning Cancer Hospital & Institute, Shenyang 110042, China.
| |
Collapse
|