1
|
Zeng Y, He Y, Wang L, Xu H, Zhang Q, Wang Y, Zhang J, Wang L. Dihydroquercetin improves experimental acute liver failure by targeting ferroptosis and mitochondria-mediated apoptosis through the SIRT1/p53 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155533. [PMID: 38552433 DOI: 10.1016/j.phymed.2024.155533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Ferroptosis and mitochondria-mediated apoptosis are both involved in the pathogenesis of acute liver failure (ALF). Ferroptosis-produced reactive oxygen species (ROS) trigger the chain oxidation of polyunsaturated phospholipids and promote mitochondrial apoptosis. Dihydroquercetin (DHQ) also plays an important protective role against liver injury. PURPOSE Here, we aimed to investigate the protective effects of DHQ on ALF. We also explored the underlying mechanism. METHODS We established a Lipopolysaccharide (LPS)/D-galactosamine (D-Gal)-induced ALF mouse model and tumor necrosis factor-α (TNF-α)/D-Gal-induced ALF LO2 cell model. 2',7'-Dichlorofluorescein diacetate (DCFH-DA) and Dihydroethidium (DHE) were used to detect total ROS levels. Lipid ROS was assessed using C11-BODIPY flow cytometry. Lipid peroxidative products levels were detected using MDA ELISA assay and 4-hydroxynonenal (4-HNE) immunohistochemistry. QRT-PCR and western blots were used to test mRNA and protein expression levels, respectively. Cell viability was evaluated with CCK8 assay, and apoptosis was analyzed using flow cytometry. RESULTS DHQ treatment improved LPS/D-Gal-induced ALF, as well as TNF-α/D-Gal-induced reductions in LO2 viability and increased sirtuin 1 (SIRT1) expression. DHQ pretreatment also reduced the accumulation of ROS, reduced lipid peroxidation, elevated mitochondrial membrane potentials (ΔΨm), and decreased liver cell apoptosis both in vivo and in vitro. Additionally, the knockdown of SIRT1 and p53 activator (Tenovin-6) treatment reversed DHQ's inhibitory effects on ferroptosis and mitochondria-mediated apoptosis in vitro. DHQ enhanced p53 deacetylation by both up-regulating SIRT1 expression and directly bonding to SIRT1. We also found that Tenovin-6's stimulatory effects on ferroptosis and mitochondria-mediated apoptosis in the DHQ-treated LO2 ALF cell model were partially attenuated by overexpression of solute carrier family 7member 11 (SLC7A11), as well as by apoptotic protease activating factor 1 (Apaf-1) knockdown. CONCLUSION Our results suggest that DHQ alleviated ALF by inhibiting both ferroptosis and mitochondria-mediated apoptosis by regulating the SIRT1/p53 axis. Thus, DHQ may serve as a novel therapy for ALF.
Collapse
Affiliation(s)
- Yuqiao Zeng
- Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Yiyu He
- Department of Cardiovascular Disease, Renmin Hospital of Wuhan University, Wuchang District, Wuhan City, Hubei 430060, China
| | - Li Wang
- Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Hao Xu
- Infection Control Center, Linyi People's Hospital. Lanshan District, Linyi City, Shandong 276000, China
| | - Qianwen Zhang
- Infection Control Center, Linyi People's Hospital. Lanshan District, Linyi City, Shandong 276000, China
| | - Yanjun Wang
- Jilin Jianwei Natural Biotechnology Co., Ltd. Kuancheng District, Changchun City, Jilin 510664, China
| | - Jianhua Zhang
- Outpatient Department, Shandong Public Health Clinical Center, Lixia District, Jinan City, Shandong 250100, China
| | - Likun Wang
- Infection Control Center, Linyi People's Hospital. Lanshan District, Linyi City, Shandong 276000, China.
| |
Collapse
|
2
|
Ladds MJGW, Popova G, Pastor-Fernández A, Kannan S, van Leeuwen IMM, Håkansson M, Walse B, Tholander F, Bhatia R, Verma CS, Lane DP, Laín S. Exploitation of dihydroorotate dehydrogenase (DHODH) and p53 activation as therapeutic targets: A case study in polypharmacology. J Biol Chem 2020; 295:17935-17949. [PMID: 32900849 PMCID: PMC7939445 DOI: 10.1074/jbc.ra119.012056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 08/18/2020] [Indexed: 01/17/2023] Open
Abstract
The tenovins are a frequently studied class of compounds capable of inhibiting sirtuin activity, which is thought to result in increased acetylation and protection of the tumor suppressor p53 from degradation. However, as we and other laboratories have shown previously, certain tenovins are also capable of inhibiting autophagic flux, demonstrating the ability of these compounds to engage with more than one target. In this study, we present two additional mechanisms by which tenovins are able to activate p53 and kill tumor cells in culture. These mechanisms are the inhibition of a key enzyme of the de novo pyrimidine synthesis pathway, dihydroorotate dehydrogenase (DHODH), and the blockage of uridine transport into cells. These findings hold a 3-fold significance: first, we demonstrate that tenovins, and perhaps other compounds that activate p53, may activate p53 by more than one mechanism; second, that work previously conducted with certain tenovins as SirT1 inhibitors should additionally be viewed through the lens of DHODH inhibition as this is a major contributor to the mechanism of action of the most widely used tenovins; and finally, that small changes in the structure of a small molecule can lead to a dramatic change in the target profile of the molecule even when the phenotypic readout remains static.
Collapse
Affiliation(s)
- Marcus J G W Ladds
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Gergana Popova
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Andrés Pastor-Fernández
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | - Fredrik Tholander
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ravi Bhatia
- Division of Hematology and Oncology, O'Neal Comprehensive Cancer Center, University of Alabama, Birmingham, Alabama, USA
| | - Chandra S Verma
- Bioinformatics Institute (BII), A*STAR, Singapore; Department of Biological Sciences, National University of Singapore, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore
| | - David P Lane
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sonia Laín
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
3
|
Abraham A, Qiu S, Chacko BK, Li H, Paterson A, He J, Agarwal P, Shah M, Welner R, Darley-Usmar VM, Bhatia R. SIRT1 regulates metabolism and leukemogenic potential in CML stem cells. J Clin Invest 2019; 129:2685-2701. [PMID: 31180336 PMCID: PMC6597223 DOI: 10.1172/jci127080] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/25/2019] [Indexed: 12/31/2022] Open
Abstract
Chronic myeloid leukemia (CML) results from hematopoietic stem cell transformation by the BCR-ABL kinase. Despite the success of BCR-ABL tyrosine kinase inhibitors (TKIs) in treating CML patients, leukemia stem cells (LSCs) resist elimination and persist as a major barrier to cure. Previous studies suggest that overexpression of the sirtuin 1 (SIRT1) deacetylase may contribute to LSC maintenance in CML. Here, by genetically deleting SIRT1 in transgenic CML mice, we definitively demonstrated an important role for SIRT1 in leukemia development. We identified a previously unrecognized role for SIRT1 in mediating increased mitochondrial oxidative phosphorylation in CML LSCs. We showed that mitochondrial alterations were kinase independent and that TKI treatment enhanced inhibition of CML hematopoiesis in SIRT1-deleted mice. We further showed that the SIRT1 substrate PGC-1α contributed to increased oxidative phosphorylation and TKI resistance in CML LSCs. These results reveal an important role for SIRT1 and downstream signaling mechanisms in altered mitochondrial respiration in CML LSCs.
Collapse
MESH Headings
- Animals
- Drug Resistance, Neoplasm
- Gene Deletion
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Mice, Transgenic
- Mitochondria/genetics
- Mitochondria/metabolism
- Mitochondria/pathology
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplastic Stem Cells/enzymology
- Neoplastic Stem Cells/pathology
- Oxygen Consumption/genetics
- Protein Kinase Inhibitors/pharmacology
- Sirtuin 1/biosynthesis
- Sirtuin 1/genetics
Collapse
Affiliation(s)
- Ajay Abraham
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Shaowei Qiu
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Balu K. Chacko
- Department of Pathology, Mitochondrial Medicine Laboratory, UAB, Birmingham, Alabama, USA
| | - Hui Li
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Andrew Paterson
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Jianbo He
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Puneet Agarwal
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Mansi Shah
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Robert Welner
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Victor M. Darley-Usmar
- Department of Pathology, Mitochondrial Medicine Laboratory, UAB, Birmingham, Alabama, USA
| | - Ravi Bhatia
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| |
Collapse
|
4
|
Ladds MJGW, Laín S. Small molecule activators of the p53 response. J Mol Cell Biol 2019; 11:245-254. [PMID: 30689917 PMCID: PMC6478124 DOI: 10.1093/jmcb/mjz006] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/21/2018] [Accepted: 01/18/2019] [Indexed: 01/10/2023] Open
Abstract
Drugging the p53 pathway has been a goal for both academics and pharmaceutical companies since the designation of p53 as the 'guardian of the genome'. Through growing understanding of p53 biology, we can see multiple routes for activation of both wild-type p53 function and restoration of mutant p53. In this review, we focus on small molecules that activate wild-type p53 and that do so in a non-genotoxic manner. In particular, we will describe potential approaches to targeting proteins that alter p53 stability and function through posttranslational modification, affect p53's subcellular localization, or target RNA synthesis or the synthesis of ribonucleotides. The plethora of pathways for exploitation of p53, as well as the wide-ranging response to p53 activation, makes it an attractive target for anti-cancer therapy.
Collapse
Affiliation(s)
- Marcus J G W Ladds
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Solnavägen 9, Karolinska Institutet, Stockholm, Sweden
- SciLifeLab, Tomtebodavägen 23A, Solna, Stockholm, Sweden
| | - Sonia Laín
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Solnavägen 9, Karolinska Institutet, Stockholm, Sweden
- SciLifeLab, Tomtebodavägen 23A, Solna, Stockholm, Sweden
| |
Collapse
|