101
|
Nasrabadi M, Nazarian M, Darroudi M, Marouzi S, Harifi-Mood MS, Samarghandian S, Farkhondeh T. Carbamate compounds induced toxic effects by affecting Nrf2 signaling pathways. Toxicol Rep 2024; 12:148-157. [PMID: 38304697 PMCID: PMC10831123 DOI: 10.1016/j.toxrep.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 02/03/2024] Open
Abstract
Carbamate (CBs) is a class of insecticides which is being known as an important cause of intentional or accidental poisoning. CBs, cause carbamylation of acetylcholinesterase at neuronal synapses and neuromuscular junction. Exposure to CBs through skin contact, inhalation, or ingestion can result in significant cholinergic toxicity. This is due to the elevation of acetylcholine levels at ganglionic synapses found in both the sympathetic and parasympathetic nervous systems, as well as muscarinic receptors located in target organs of the parasympathetic nervous system, nicotinic receptors situated in skeletal muscle tissue, and the central nervous system. The association between human illnesses and environmental exposures to CBs have been extensively studied in several studies. Although CBs-triggered toxicity leads to overproduction of reactive oxygen species (ROS), the detailed association between the toxicity under CBs exposure and NFE2-related factor 2 (Nrf2) signaling pathways has not been completely clarified. In this review we aimed to summarize the latest findings on the functional interrelationship between carbamates compounds and Nrf2 signaling.
Collapse
Affiliation(s)
| | - Maryam Nazarian
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Majid Darroudi
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Basic Sciences, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran
| | - Somayeh Marouzi
- Department of Basic Sciences, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad, Iran
| | | | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran
| | - Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
102
|
Luo YM, Liu SS, Zhao M, Wei W, Yao JX, Sun JH, Cao Y, Li H. Crosstalk among Oxidative Stress, Autophagy, and Apoptosis in the Protective Effects of Ginsenoside Rb1 on Brain Microvascular Endothelial Cells: A Mixed Computational and Experimental Study. Curr Med Sci 2024; 44:578-588. [PMID: 38853191 DOI: 10.1007/s11596-024-2858-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/28/2024] [Indexed: 06/11/2024]
Abstract
OBJECTIVE Brain microvascular endothelial cells (BMECs) were found to shift from their usually inactive state to an active state in ischemic stroke (IS) and cause neuronal damage. Ginsenoside Rb1 (GRb1), a component derived from medicinal plants, is known for its pharmacological benefits in IS, but its protective effects on BMECs have yet to be explored. This study aimed to investigate the potential protective effects of GRb1 on BMECs. METHODS An in vitro oxygen-glucose deprivation/reperfusion (OGD/R) model was established to mimic ischemia-reperfusion (I/R) injury. Bulk RNA-sequencing data were analyzed by using the Human Autophagy Database and various bioinformatic tools, including gene set enrichment analysis (GSEA), Gene Ontology (GO) classification and enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, protein-protein interaction network analysis, and molecular docking. Experimental validation was also performed to ensure the reliability of our findings. RESULTS Rb1 had a protective effect on BMECs subjected to OGD/R injury. Specifically, GRb1 was found to modulate the interplay between oxidative stress, apoptosis, and autophagy in BMECs. Key targets such as sequestosome 1 (SQSTM1/p62), autophagy related 5 (ATG5), and hypoxia-inducible factor 1-alpha (HIF-1α) were identified, highlighting their potential roles in mediating the protective effects of GRb1 against IS-induced damage. CONCLUSION GRbl protects BMECs against OGD/R injury by influencing oxidative stress, apoptosis, and autophagy. The identification of SQSTM1/p62, ATG5, and HIF-1α as promising targets further supports the potential of GRb1 as a therapeutic agent for IS, providing a foundation for future research into its mechanisms and applications in IS treatment.
Collapse
Affiliation(s)
- Yi-Miao Luo
- Department of Geriatrics, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, 100901, China
- Department of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Shu-Sen Liu
- School of Pharmacy, Harbin University of Commerce, Harbin, 150028, China
| | - Ming Zhao
- Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, 100901, China
| | - Wei Wei
- Wangjing Hospital, China Academy of Chinese Medical Science, Beijing, 100102, China
| | - Jiu-Xiu Yao
- Wangjing Hospital, China Academy of Chinese Medical Science, Beijing, 100102, China
| | - Jia-Hui Sun
- Wangjing Hospital, China Academy of Chinese Medical Science, Beijing, 100102, China
| | - Yu Cao
- Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, 100901, China.
| | - Hao Li
- Department of Geriatrics, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, 100901, China.
- Department of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, 100191, China.
- Wangjing Hospital, China Academy of Chinese Medical Science, Beijing, 100102, China.
| |
Collapse
|
103
|
Balhara M, Neikirk K, Marshall A, Hinton A, Kirabo A. Endoplasmic Reticulum Stress in Hypertension and Salt Sensitivity of Blood Pressure. Curr Hypertens Rep 2024; 26:273-290. [PMID: 38602583 PMCID: PMC11166838 DOI: 10.1007/s11906-024-01300-9] [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] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
PURPOSE OF REVIEW Hypertension is a principal risk factor for cardiovascular morbidity and mortality, with its severity exacerbated by high sodium intake, particularly in individuals with salt-sensitive blood pressure. However, the mechanisms underlying hypertension and salt sensitivity are only partly understood. Herein, we review potential interactions in hypertension pathophysiology involving the immune system, endoplasmic reticulum (ER) stress, the unfolded protein response (UPR), and proteostasis pathways; identify knowledge gaps; and discuss future directions. RECENT FINDINGS Recent advancements by our research group and others reveal interactions within and between adaptive and innate immune responses in hypertension pathophysiology. The salt-immune-hypertension axis is further supported by the discovery of the role of dendritic cells in hypertension, marked by isolevuglandin (IsoLG) formation. Alongside these broadened understandings of immune-mediated salt sensitivity, the contributions of T cells to hypertension have been recently challenged by groups whose findings did not support increased resistance of Rag-1-deficient mice to Ang II infusion. Hypertension has also been linked to ER stress and the UPR. Notably, a holistic approach is needed because the UPR engages in crosstalk with autophagy, the ubiquitin proteasome, and other proteostasis pathways, that may all involve hypertension. There is a critical need for studies to establish cause and effect relationships between ER stress and the UPR in hypertension pathophysiology in humans and to determine whether the immune system and ER stress function mainly to exacerbate or initiate hypertension and target organ injury. This review of recent studies proposes new avenues for future research for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Maria Balhara
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Andrea Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA.
- Vanderbilt Center for Immunobiology, Nashville, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, USA.
- Vanderbilt Institute for Global Health, Nashville, USA.
| |
Collapse
|
104
|
An N, Wang R, Li L, Wang B, Wang H, Peng G, Zhou H, Chen G. Celastrol alleviates diabetic vascular injury via Keap1/Nrf2-mediated anti-inflammation. Front Pharmacol 2024; 15:1360177. [PMID: 38881873 PMCID: PMC11176472 DOI: 10.3389/fphar.2024.1360177] [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: 12/22/2023] [Accepted: 05/10/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction: Celastrol (Cel) is a widely used main component of Chinese herbal medicine with strong anti-inflammatory, antiviral and antitumor activities. In the present study, we aimed to elucidate the cellular molecular protective mechanism of Cel against diabetes-induced inflammation and endothelial dysfunction. Methods: Type 2 diabetes (T2DM) was induced by db/db mice, and osmotic pumps containing Cel (100 μg/kg/day) were implanted intraperitoneally and were calibrated to release the drug for 28 days. In addition, human umbilical vein endothelial cells (HUVECs) were cultured in normal or high glucose and palmitic acid-containing (HG + PA) media in the presence or absence of Cel for 48 h. Results: Cel significantly ameliorated the hyperglycemia-induced abnormalities in nuclear factor (erythroid-derived 2)-like protein 2 (Nrf2) pathway activity and alleviated HG + PA-induced oxidative damage. However, the protective effect of Cel was almost completely abolished in HUVECs transfected with short hairpin (sh)RNA targeting Nrf2, but not by nonsense shRNA. Furthermore, HG + PA reduced the phosphorylation of AMP-activated protein kinase (AMPK), the autophagic degradation of p62/Kelch-like ECH-associated protein 1 (Keap1), and the nuclear localization of Nrf2. However, these catabolic pathways were inhibited by Cel treatment in HUVECs. In addition, compound C (AMPK inhibitors) and AAV9-sh-Nrf2 reduced Cel-induced Nrf2 activation and angiogenesis in db/db mice. Discussion: Taking these findings together, the endothelial protective effect of Cel in the presence of HG + PA may be at least in part attributed to its effects to reduce reactive oxygen species (ROS) and inflammation through p62/Keap1-mediated Nrf2 activation.
Collapse
Affiliation(s)
- Ning An
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
| | - Rixiang Wang
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
| | - Lin Li
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
| | - Bingyu Wang
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
| | - Huiting Wang
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, China
| | - Ganyu Peng
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, China
| | - Hua Zhou
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
| | - Gen Chen
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
105
|
Iibushi J, Nozawa T, Toh H, Nakagawa I. ATG9B regulates bacterial internalization via actin rearrangement. iScience 2024; 27:109623. [PMID: 38706859 PMCID: PMC11066431 DOI: 10.1016/j.isci.2024.109623] [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: 10/10/2023] [Revised: 01/16/2024] [Accepted: 03/26/2024] [Indexed: 05/07/2024] Open
Abstract
Invasive bacterial pathogens are internalized by host cells through endocytosis, which is regulated by a cascade of actin rearrangement signals triggered by host cell receptors or bacterial proteins delivered into host cells. However, the molecular mechanisms that mediate actin rearrangement to promote bacterial invasion are not fully understood. Here, we show that the autophagy-related (ATG) protein ATG9B regulates the internalization of various bacteria by controlling actin rearrangement. ATG knockout screening and knockdown experiments in HeLa cells identified ATG9B as a critical factor for bacterial internalization. In particular, cells with ATG9B knockdown exhibited an accumulation of actin filaments and phosphorylated LIM kinase and cofilin, suggesting that ATG9B is involved in actin depolymerization. Furthermore, the kinase activity of Unc-51-like autophagy-activating kinase 1 was found to regulate ATG9B localization and actin remodeling. These findings revealed a newly discovered function of ATG proteins in bacterial infection rather than autophagy-mediated immunity.
Collapse
Affiliation(s)
- Junpei Iibushi
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| | - Takashi Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| | - Hirotaka Toh
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| |
Collapse
|
106
|
Gorska-Arcisz M, Popeda M, Braun M, Piasecka D, Nowak JI, Kitowska K, Stasilojc G, Okroj M, Romanska HM, Sadej R. FGFR2-triggered autophagy and activation of Nrf-2 reduce breast cancer cell response to anti-ER drugs. Cell Mol Biol Lett 2024; 29:71. [PMID: 38745155 PMCID: PMC11092031 DOI: 10.1186/s11658-024-00586-6] [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: 11/29/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Genetic abnormalities in the FGFR signalling occur in 40% of breast cancer (BCa) patients resistant to anti-ER therapy, which emphasizes the potential of FGFR-targeting strategies. Recent findings indicate that not only mutated FGFR is a driver of tumour progression but co-mutational landscapes and other markers should be also investigated. Autophagy has been recognized as one of the major mechanisms underlying the role of tumour microenvironment in promotion of cancer cell survival, and resistance to anti-ER drugs. The selective autophagy receptor p62/SQSTM1 promotes Nrf-2 activation by Keap1/Nrf-2 complex dissociation. Herein, we have analysed whether the negative effect of FGFR2 on BCa cell response to anti-ER treatment involves the autophagy process and/or p62/Keap1/Nrf-2 axis. METHODS The activity of autophagy in ER-positive MCF7 and T47D BCa cell lines was determined by analysis of expression level of autophagy markers (p62 and LC3B) and monitoring of autophagosomes' maturation. Western blot, qPCR and proximity ligation assay were used to determine the Keap1/Nrf-2 interaction and Nrf-2 activation. Analysis of 3D cell growth in Matrigel® was used to assess BCa cell response to applied treatments. In silico gene expression analysis was performed to determine FGFR2/Nrf-2 prognostic value. RESULTS We have found that FGFR2 signalling induced autophagy in AMPKα/ULK1-dependent manner. FGFR2 activity promoted dissociation of Keap1/Nrf-2 complex and activation of Nrf-2. Both, FGFR2-dependent autophagy and activation of Nrf-2 were found to counteract the effect of anti-ER drugs on BCa cell growth. Moreover, in silico analysis showed that high expression of NFE2L2 (gene encoding Nrf-2) combined with high FGFR2 expression was associated with poor relapse-free survival (RFS) of ER+ BCa patients. CONCLUSIONS This study revealed the unknown role of FGFR2 signalling in activation of autophagy and regulation of the p62/Keap1/Nrf-2 interdependence, which has a negative impact on the response of ER+ BCa cells to anti-ER therapies. The data from in silico analyses suggest that expression of Nrf-2 could act as a marker indicating potential benefits of implementation of anti-FGFR therapy in patients with ER+ BCa, in particular, when used in combination with anti-ER drugs.
Collapse
Affiliation(s)
- Monika Gorska-Arcisz
- Laboratory of Enzymology and Molecular Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Marta Popeda
- Department of Pathomorphology, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Braun
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Pomorska 251, 92-213, Lodz, Poland
| | - Dominika Piasecka
- Laboratory of Enzymology and Molecular Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Joanna I Nowak
- Department of Histology, Medical University of Gdansk, Gdansk, Poland
| | - Kamila Kitowska
- Laboratory of Enzymology and Molecular Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Grzegorz Stasilojc
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Marcin Okroj
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Hanna M Romanska
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Pomorska 251, 92-213, Lodz, Poland.
| | - Rafal Sadej
- Laboratory of Enzymology and Molecular Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland.
| |
Collapse
|
107
|
Deja S, Fletcher JA, Kim CW, Kucejova B, Fu X, Mizerska M, Villegas M, Pudelko-Malik N, Browder N, Inigo-Vollmer M, Menezes CJ, Mishra P, Berglund ED, Browning JD, Thyfault JP, Young JD, Horton JD, Burgess SC. Hepatic malonyl-CoA synthesis restrains gluconeogenesis by suppressing fat oxidation, pyruvate carboxylation, and amino acid availability. Cell Metab 2024; 36:1088-1104.e12. [PMID: 38447582 PMCID: PMC11081827 DOI: 10.1016/j.cmet.2024.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 12/10/2023] [Accepted: 02/09/2024] [Indexed: 03/08/2024]
Abstract
Acetyl-CoA carboxylase (ACC) promotes prandial liver metabolism by producing malonyl-CoA, a substrate for de novo lipogenesis and an inhibitor of CPT-1-mediated fat oxidation. We report that inhibition of ACC also produces unexpected secondary effects on metabolism. Liver-specific double ACC1/2 knockout (LDKO) or pharmacologic inhibition of ACC increased anaplerosis, tricarboxylic acid (TCA) cycle intermediates, and gluconeogenesis by activating hepatic CPT-1 and pyruvate carboxylase flux in the fed state. Fasting should have marginalized the role of ACC, but LDKO mice maintained elevated TCA cycle intermediates and preserved glycemia during fasting. These effects were accompanied by a compensatory induction of proteolysis and increased amino acid supply for gluconeogenesis, which was offset by increased protein synthesis during feeding. Such adaptations may be related to Nrf2 activity, which was induced by ACC inhibition and correlated with fasting amino acids. The findings reveal unexpected roles for malonyl-CoA synthesis in liver and provide insight into the broader effects of pharmacologic ACC inhibition.
Collapse
Affiliation(s)
- Stanislaw Deja
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Justin A Fletcher
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Chai-Wan Kim
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Blanka Kucejova
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Xiaorong Fu
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Monika Mizerska
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Morgan Villegas
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Natalia Pudelko-Malik
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Nicholas Browder
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Melissa Inigo-Vollmer
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Cameron J Menezes
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Prashant Mishra
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Eric D Berglund
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Jeffrey D Browning
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - John P Thyfault
- Departments of Cell Biology and Physiology, Internal Medicine and KU Diabetes Institute, Kansas Medical Center, Kansas City, KS, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| | - Jay D Horton
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA.
| | - Shawn C Burgess
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA.
| |
Collapse
|
108
|
Diao J, Jia Y, Dai E, Liu J, Kang R, Tang D, Han L, Zhong Y, Meng L. Ferroptotic therapy in cancer: benefits, side effects, and risks. Mol Cancer 2024; 23:89. [PMID: 38702722 PMCID: PMC11067110 DOI: 10.1186/s12943-024-01999-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/10/2024] [Indexed: 05/06/2024] Open
Abstract
Ferroptosis is a type of regulated cell death characterized by iron accumulation and uncontrolled lipid peroxidation, leading to plasma membrane rupture and intracellular content release. Originally investigated as a targeted therapy for cancer cells carrying oncogenic RAS mutations, ferroptosis induction now exhibits potential to complement chemotherapy, immunotherapy, and radiotherapy in various cancer types. However, it can lead to side effects, including immune cell death, bone marrow impairment, liver and kidney damage, cachexia (severe weight loss and muscle wasting), and secondary tumorigenesis. In this review, we discuss the advantages and offer an overview of the diverse range of documented side effects. Furthermore, we examine the underlying mechanisms and explore potential strategies for side effect mitigation.
Collapse
Affiliation(s)
- Jiandong Diao
- 2nd Inpatient Area of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China
| | - Yuanyuan Jia
- 2nd Inpatient Area of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China
| | - Enyong Dai
- 2nd Inpatient Area of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China
| | - Jiao Liu
- DAMP laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Rui Kang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daolin Tang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Leng Han
- 2nd Inpatient Area of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China.
| | - Yingjie Zhong
- Department of Pediatrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China.
| | - Lingjun Meng
- 2nd Inpatient Area of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China.
| |
Collapse
|
109
|
Wang YY, Ni JC, Zhao YQ, Yang X, Niu ZP, Yang XZ, Dong XX, Zhao YH, Hao XJ, Ding X. Iridoid glycosides from Morinda officinalis induce lysosomal biogenesis and promote autophagic flux to attenuate oxidative stress. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:562-574. [PMID: 37897053 DOI: 10.1080/10286020.2023.2269370] [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: 03/06/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023]
Abstract
Morinda officinalis is a traditional Chinese tonic herb, and have been used in the treatment of multiple diseases. Here, three iridoid glycosides isolated from M. officinalis were evaluated for their roles in the autophagy-lysosomal pathway. All three iridoid glycosides could induce TFEB/TFE3-mediated lysosomal biogenesis and trigger autophagy. Interestingly, they promoted the nuclear import of TFEB/TFE3 without affecting their nuclear export, suggesting their role in the maintenance of lysosomal homeostasis. The results from this study shed light on the identification of autophagy activators from M. officinalis and provide a basis for developing them in the treatment of oxidative stress-involved diseases.
Collapse
Affiliation(s)
- Yin-Yuan Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Jian-Cheng Ni
- The Engineering Technology Research Center of Characteristic Medicinal Plants of Fujian, Ningde Normal University, Ningde 352100, China
| | - Yue-Qin Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen-Peng Niu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing 100730, China
- School of Basic Medicine, Guizhou Medical University, Guiyang 550009, China
| | - Xing-Zhi Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Xian-Xiang Dong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu-Han Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Xiao-Jiang Hao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiao Ding
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
110
|
Ghosh R, Fatahian AN, Rouzbehani OMT, Hathaway MA, Mosleh T, Vinod V, Vowles S, Stephens SL, Chung SLD, Cao ID, Jonnavithula A, Symons JD, Boudina S. Sequestosome 1 (p62) mitigates hypoxia-induced cardiac dysfunction by stabilizing hypoxia-inducible factor 1α and nuclear factor erythroid 2-related factor 2. Cardiovasc Res 2024; 120:531-547. [PMID: 38332738 PMCID: PMC11060490 DOI: 10.1093/cvr/cvae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 09/11/2023] [Accepted: 11/03/2023] [Indexed: 02/10/2024] Open
Abstract
AIMS Heart failure due to ischaemic heart disease (IHD) is a leading cause of mortality worldwide. A major contributing factor to IHD-induced cardiac damage is hypoxia. Sequestosome 1 (p62) is a multi-functional adaptor protein with pleiotropic roles in autophagy, proteostasis, inflammation, and cancer. Despite abundant expression in cardiomyocytes, the role of p62 in cardiac physiology is not well understood. We hypothesized that cardiomyocyte-specific p62 deletion evokes hypoxia-induced cardiac pathology by impairing hypoxia-inducible factor 1α (Hif-1α) and nuclear factor erythroid 2-related factor 2 (Nrf2) signalling. METHODS AND RESULTS Adult mice with germline deletion of cardiomyocyte p62 exhibited mild cardiac dysfunction under normoxic conditions. Transcriptomic analyses revealed a selective impairment in Nrf2 target genes in the hearts from these mice. Demonstrating the functional importance of this adaptor protein, adult mice with inducible depletion of cardiomyocyte p62 displayed hypoxia-induced contractile dysfunction, oxidative stress, and cell death. Mechanistically, p62-depleted hearts exhibit impaired Hif-1α and Nrf2 transcriptional activity. Because findings from these two murine models suggested a cardioprotective role for p62, mechanisms were evaluated using H9c2 cardiomyoblasts. Loss of p62 in H9c2 cells exposed to hypoxia reduced Hif-1α and Nrf2 protein levels. Further, the lack of p62 decreased Nrf2 protein expression, nuclear translocation, and transcriptional activity. Repressed Nrf2 activity associated with heightened Nrf2-Keap1 co-localization in p62-deficient cells, which was concurrent with increased Nrf2 ubiquitination facilitated by the E3 ligase Cullin 3, followed by proteasomal-mediated degradation. Substantiating our results, a gain of p62 in H9c2 cells stabilized Nrf2 and increased the transcriptional activity of Nrf2 downstream targets. CONCLUSION Cardiac p62 mitigates hypoxia-induced cardiac dysfunction by stabilizing Hif-1α and Nrf2.
Collapse
Affiliation(s)
- Rajeshwary Ghosh
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program (U2M2), University of Utah, Salt Lake City, UT 84112, USA
| | - Amir Nima Fatahian
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - Omid M T Rouzbehani
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - Marissa A Hathaway
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - Tariq Mosleh
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - Vishaka Vinod
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - Sidney Vowles
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - Sophie L Stephens
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - Siu-Lai Desmond Chung
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - Isaac D Cao
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - Anila Jonnavithula
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program (U2M2), University of Utah, Salt Lake City, UT 84112, USA
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program (U2M2), University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
111
|
Liao H, Wang Y, Zou L, Fan Y, Wang X, Tu X, Zhu Q, Wang J, Liu X, Dong C. Relationship of mTORC1 and ferroptosis in tumors. Discov Oncol 2024; 15:107. [PMID: 38583115 PMCID: PMC10999401 DOI: 10.1007/s12672-024-00954-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 03/28/2024] [Indexed: 04/08/2024] Open
Abstract
Ferroptosis is a novel form of programmed death, dependent on iron ions and oxidative stress, with a predominant intracellular form of lipid peroxidation. In recent years, ferroptosis has gained more and more interest of people in the treatment mechanism of targeted tumors. mTOR, always overexpressed in the tumor, and controlling cell growth and metabolic activities, has an important role in both autophagy and ferroptosis. Interestingly, the selective types of autophay plays an important role in promoting ferroptosis, which is related to mTOR and some metabolic pathways (especially in iron and amino acids). In this paper, we list the main mechanisms linking ferroptosis with mTOR signaling pathway and further summarize the current compounds targeting ferroptosis in these ways. There are growing experimental evidences that targeting mTOR and ferroptosis may have effective impact in many tumors, and understanding the mechanisms linking mTOR to ferroptosis could provide a potential therapeutic approach for tumor treatment.
Collapse
Affiliation(s)
- Huilin Liao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Yueqing Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Lili Zou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Yanmei Fan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Xinyue Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Xiancong Tu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Qiaobai Zhu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Jun Wang
- The People's Hospital of China Three Gorges University and The First People's Hospital of Yichang, Yichang, Hubei, China, 443002
| | - Xiaowen Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002.
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China, 443002.
| | - Chuanjiang Dong
- Department of Urology, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China, 523000.
| |
Collapse
|
112
|
Yu S, Tong L, Shen J, Li C, Hu Y, Feng K, Shao J. Recent research progress based on ferroptosis-related signaling pathways and the tumor microenvironment on it effects. Eur J Med Chem 2024; 269:116290. [PMID: 38518522 DOI: 10.1016/j.ejmech.2024.116290] [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: 12/18/2023] [Revised: 02/19/2024] [Accepted: 02/25/2024] [Indexed: 03/24/2024]
Abstract
The existing therapies for cancer are not remote satisfactory due to drug-resistance in tumors that are malignant. There is a pressing necessity to take a step forward to develop innovative therapies that can complement current ones. Multiple investigations have demonstrated that ferroptosis therapy, a non-apoptotic modality of programmed cell death, has tremendous potential in face of multiple crucial events, such as drug resistance and toxicity in aggressive malignancies. Recently, ferroptosis at the crosswalk of chemotherapy, materials science, immunotherapy, tumor microenvironment, and bionanotechnology has been presented to elucidate its therapeutic feasibility. Given the burgeoning progression of ferroptosis-based nanomedicine, the newest advancements in this field at the confluence of ferroptosis-inducers, nanotherapeutics, along with tumor microenvironment are given an overview. Here, the signaling pathways of ferroptosis-related were first talked about briefly. The emphasis discussion was placed on the pharmacological mechanisms and the nanodrugs design of ferroptosis inducing agents based on multiple distinct metabolism pathways. Additionally, a comprehensive overview of the action mechanisms by which the tumor microenvironment influences ferroptosis was elaborately descripted. Finally, some limitations of current researches and future research directions were also deliberately discussed to provide details about therapeutic avenues for ferroptosis-related diseases along with the design of anti-drugs.
Collapse
Affiliation(s)
- Shijing Yu
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Lingwu Tong
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jiangwen Shen
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Chenglei Li
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yongshan Hu
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Keke Feng
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jingwei Shao
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China.
| |
Collapse
|
113
|
Laguna JG, Freitas ADS, Barroso FAL, De Jesus LCL, De Vasconcelos OAGG, Quaresma LS, Américo MF, Campos GM, Glória RDA, Dutra JDCF, Da Silva TF, Vital KD, Fernandes SO, Souza RO, Martins FDS, Ferreira E, Santos TM, Birbrair A, De Oliveira MFA, Faria AMC, Carvalho RDDO, Venanzi FM, Le Loir Y, Jan G, Guédon É, Azevedo VADC. Recombinant probiotic Lactococcus lactis delivering P62 mitigates moderate colitis in mice. Front Microbiol 2024; 15:1309160. [PMID: 38680913 PMCID: PMC11047439 DOI: 10.3389/fmicb.2024.1309160] [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: 10/07/2023] [Accepted: 02/21/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction and objective p62 is a human multifunctional adaptor protein involved in key cellular processes such as tissue homeostasis, inflammation, and cancer. It acts as a negative regulator of inflammasome complexes. It may thus be considered a good candidate for therapeutic use in inflammatory bowel diseases (IBD), such as colitis. Probiotics, including recombinant probiotic strains producing or delivering therapeutic biomolecules to the host mucosal surfaces, could help prevent and mitigate chronic intestinal inflammation. The objective of the present study was to combine the intrinsic immunomodulatory properties of the probiotic Lactococcus lactis NCDO2118 with its ability to deliver health-promoting molecules to enhance its protective and preventive effects in the context of ulcerative colitis (UC). Material and methods This study was realized in vivo in which mice were supplemented with the recombinant strain. The intestinal barrier function was analyzed by monitoring permeability, secretory IgA total levels, mucin expression, and tight junction genes. Its integrity was evaluated by histological analyses. Regarding inflammation, colonic cytokine levels, myeloperoxidase (MPO), and expression of key genes were monitored. The intestinal microbiota composition was investigated using 16S rRNA Gene Sequencing. Results and discussion No protective effect of L. lactis NCDO2118 pExu:p62 was observed regarding mice clinical parameters compared to the L. lactis NCDO2118 pExu: empty. However, the recombinant strain, expressing p62, increased the goblet cell counts, upregulated Muc2 gene expression in the colon, and downregulated pro-inflammatory cytokines Tnf and Ifng when compared to L. lactis NCDO2118 pExu: empty and inflamed groups. This recombinant strain also decreased colonic MPO activity. No difference in the intestinal microbiota was observed between all treatments. Altogether, our results show that recombinant L. lactis NCDO2118 delivering p62 protein protected the intestinal mucosa and mitigated inflammatory damages caused by dextran sodium sulfate (DSS). We thus suggest that p62 may constitute part of a therapeutic approach targeting inflammation.
Collapse
Affiliation(s)
- Juliana Guimarães Laguna
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria dos Santos Freitas
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Luís Cláudio Lima De Jesus
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Ludmila Silva Quaresma
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Américo
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gabriela Munis Campos
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rafael de Assis Glória
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Joyce da Cruz Ferraz Dutra
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Tales Fernando Da Silva
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Kátia Duarte Vital
- Department of Clinical Analysis and Toxicology, Federal University of Minas Gerais Belo Horizonte, Minas Gerais, Brazil
| | - Simone O. Fernandes
- Department of Clinical Analysis and Toxicology, Federal University of Minas Gerais Belo Horizonte, Minas Gerais, Brazil
| | - Ramon O. Souza
- Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Enio Ferreira
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Túlio Marcos Santos
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alexander Birbrair
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Ana Maria Caetano Faria
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Franco Maria Venanzi
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | | | | | | | | |
Collapse
|
114
|
Cui J, Zhu M, Sun X, Yang J, Guo M. Microplastics induced endoplasmic reticulum stress to format an inflammation and cell death in hepatocytes of carp (Cyprinus carpio). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 269:106870. [PMID: 38395010 DOI: 10.1016/j.aquatox.2024.106870] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 02/25/2024]
Abstract
Microplastics (MPs) are a serious threat to the living environment of aquatic organisms. However, there are fewer studies on the toxicity of microplastics to freshwater organisms. This study aimed to establish a polystyrene microplastics (PS-MPs) model by feeding carp (Cyprinus carpio) PS-MP (1000 ng/L) particles 8 μm in size. HE staining revealed a mass of inflammatory cells infiltrated in the carp hepatopancreas. The activities of alkaline phosphatase (AKP), aspartate transaminase (AST), lactate dehydrogenase (LDH), and alanine transaminase (ALT) were strengthened considerably, suggesting that PS-MPs cause injury to the hepatopancreas of carp. Real-Time polymerase chain reaction and western blotting results indicated increased levels of glucose-regulated protein 78 (GRP78), (PKR)-like ER kinase (PERK), eukaryotic translation initiation Factor 2α (EIF2α) and activating transcription Factor 4 (ATF4) genes and increased levels of inflammatory factors downstream of endoplasmic reticulum stress (ERs) thioredoxin-interacting protein (TXNIP), NOD-like receptor protein 3 (NLRP3), interleukin-18 (IL-18), interleukin-1β (IL-1β), and caspase 1. Increased expression of microtubule-associated protein-2 (LC3II), autophagy-related 5 (ATG5) and autophagy-related 12 (ATG12) genes revealed that PS-MPs promoted autophagy in carp hepatocytes. The enhanced expression of the Caspase 12, Caspase 3, and Bax genes suggested that PS-MPs led to the apoptosis of carp hepatocytes. These results suggest that PS-MPs result in serious injury to the hepatopancreas of carp. The present study of PS-MPs in freshwater fish from the aspect of endoplasmic reticulum stress was conducted to provide references and suggestions for toxicological studies of PS-MPs in freshwater environments.
Collapse
Affiliation(s)
- Jie Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Mengran Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaoran Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jie Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Mengyao Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
115
|
Kowalik MA, Taguchi K, Serra M, Caddeo A, Puliga E, Bacci M, Koshiba S, Inoue J, Hishinuma E, Morandi A, Giordano S, Perra A, Yamamoto M, Columbano A. Metabolic reprogramming in Nrf2-driven proliferation of normal rat hepatocytes. Hepatology 2024; 79:829-843. [PMID: 37603610 DOI: 10.1097/hep.0000000000000568] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 07/31/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND AND AIMS Cancer cells reprogram their metabolic pathways to support bioenergetic and biosynthetic needs and to maintain their redox balance. In several human tumors, the Keap1-Nrf2 system controls proliferation and metabolic reprogramming by regulating the pentose phosphate pathway (PPP). However, whether this metabolic reprogramming also occurs in normal proliferating cells is unclear. APPROACH AND RESULTS To define the metabolic phenotype in normal proliferating hepatocytes, we induced cell proliferation in the liver by 3 distinct stimuli: liver regeneration by partial hepatectomy and hepatic hyperplasia induced by 2 direct mitogens: lead nitrate (LN) or triiodothyronine. Following LN treatment, well-established features of cancer metabolic reprogramming, including enhanced glycolysis, oxidative PPP, nucleic acid synthesis, NAD + /NADH synthesis, and altered amino acid content, as well as downregulated oxidative phosphorylation, occurred in normal proliferating hepatocytes displaying Nrf2 activation. Genetic deletion of Nrf2 blunted LN-induced PPP activation and suppressed hepatocyte proliferation. Moreover, Nrf2 activation and following metabolic reprogramming did not occur when hepatocyte proliferation was induced by partial hepatectomy or triiodothyronine. CONCLUSIONS Many metabolic changes in cancer cells are shared by proliferating normal hepatocytes in response to a hostile environment. Nrf2 activation is essential for bridging metabolic changes with crucial components of cancer metabolic reprogramming, including the activation of oxidative PPP. Our study demonstrates that matured hepatocytes exposed to LN undergo cancer-like metabolic reprogramming and offers a rapid and useful in vivo model to study the molecular alterations underpinning the differences/similarities of metabolic changes in normal and neoplastic hepatocytes.
Collapse
Affiliation(s)
- Marta A Kowalik
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Keiko Taguchi
- Department of Molecular Biology and Biochemistry, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Advanced Research Center for Innovations in Next Generation Medicine (INGEM), Tohoku University, Sendai, Japan
| | - Marina Serra
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Andrea Caddeo
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Elisabetta Puliga
- Department of Oncology, University of Torino, Candiolo, Italy
- Department of Oncology Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Marina Bacci
- Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Florence, Italy
| | - Seizo Koshiba
- Department of Molecular Biology and Biochemistry, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Advanced Research Center for Innovations in Next Generation Medicine (INGEM), Tohoku University, Sendai, Japan
| | - Jin Inoue
- Department of Molecular Biology and Biochemistry, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Advanced Research Center for Innovations in Next Generation Medicine (INGEM), Tohoku University, Sendai, Japan
| | - Eiji Hishinuma
- Department of Molecular Biology and Biochemistry, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Advanced Research Center for Innovations in Next Generation Medicine (INGEM), Tohoku University, Sendai, Japan
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Florence, Italy
| | - Silvia Giordano
- Department of Oncology, University of Torino, Candiolo, Italy
- Department of Oncology Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Andrea Perra
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Masayuki Yamamoto
- Department of Molecular Biology and Biochemistry, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Advanced Research Center for Innovations in Next Generation Medicine (INGEM), Tohoku University, Sendai, Japan
| | - Amedeo Columbano
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| |
Collapse
|
116
|
Zou T, Xie R, Huang S, Lu D, Liu J. Potential role of modulating autophagy levels in sensorineural hearing loss. Biochem Pharmacol 2024; 222:116115. [PMID: 38460910 DOI: 10.1016/j.bcp.2024.116115] [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: 01/14/2024] [Revised: 02/20/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
In recent years, extensive research has been conducted on the pathogenesis of sensorineural hearing loss (SNHL). Apoptosis and necrosis have been identified to play important roles in hearing loss, but they cannot account for all hearing loss. Autophagy, a cellular process responsible for cell self-degradation and reutilization, has emerged as a significant factor contributing to hearing loss, particularly in cases of autophagy deficiency. Autophagy plays a crucial role in maintaining cell health by exerting cytoprotective and metabolically homeostatic effects in organisms. Consequently, modulating autophagy levels can profoundly impact the survival, death, and regeneration of cells in the inner ear, including hair cells (HCs) and spiral ganglion neurons (SGNs). Abnormal mitochondrial autophagy has been demonstrated in animal models of SNHL. These findings indicate the profound significance of comprehending autophagy while suggesting that our perspective on this cellular process holds promise for advancing the treatment of SNHL. Thus, this review aims to clarify the pathogenic mechanisms of SNHL and the role of autophagy in the developmental processes of various cochlear structures, including the greater epithelial ridge (GER), SGNs, and the ribbon synapse. The pathogenic mechanisms of age-related hearing loss (ARHL), also known as presbycusis, and the latest research on autophagy are also discussed. Furthermore, we underscore recent findings on the modulation of autophagy in SNHL induced by ototoxic drugs. Additionally, we suggest further research that might illuminate the complete potential of autophagy in addressing SNHL, ultimately leading to the formulation of pioneering therapeutic strategies and approaches for the treatment of deafness.
Collapse
Affiliation(s)
- Ting Zou
- Department of Otorhinolaryngology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Renwei Xie
- Department of Otorhinolaryngology, Renhe Hospital, Baoshan District, Shanghai, China
| | - Sihan Huang
- Department of Otorhinolaryngology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dingkun Lu
- Cardiac Arrhythmia Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Liu
- Department of Otorhinolaryngology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
117
|
Luo J, Li P, Dong M, Zhang Y, Lu S, Chen M, Zhou H, Lin N, Jiang H, Wang Y. SLC15A3 plays a crucial role in pulmonary fibrosis by regulating macrophage oxidative stress. Cell Death Differ 2024; 31:417-430. [PMID: 38374230 PMCID: PMC11043330 DOI: 10.1038/s41418-024-01266-w] [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: 08/24/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/21/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal and irreversible disease with few effective treatments. Alveolar macrophages (AMs) are involved in the development of IPF from the initial stages due to direct exposure to air and respond to external oxidative damage (a major inducement of pulmonary fibrosis). Oxidative stress in AMs plays an indispensable role in promoting fibrosis development. The oligopeptide histidine transporter SLC15A3, mainly expressed on the lysosomal membrane of macrophages and highly expressed in the lung, has proved to be involved in innate immune and antiviral signaling pathways. In this study, we demonstrated that during bleomycin (BLM)- or radiation-induced pulmonary fibrosis, the recruitment of macrophages induced an increase of SLC15A3 in the lung, and the deficiency of SLC15A3 protected mice from pulmonary fibrosis and maintained the homeostasis of the pulmonary microenvironment. Mechanistically, deficiency of SLC15A3 resisted oxidative stress in macrophages, and SLC15A3 interacted with the scaffold protein p62 to regulate its expression and phosphorylation activation, thereby regulating p62-nuclear factor erythroid 2-related factor 2 (NRF2) antioxidant stress pathway protein, which is related to the production of reactive oxygen species (ROS). Overall, our data provided a novel mechanism for targeting SLC15A3 to regulate oxidative stress in macrophages, supporting the therapeutic potential of inhibiting or silencing SLC15A3 for the precautions and treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jun Luo
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ping Li
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China
| | - Minlei Dong
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yingqiong Zhang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shuanghui Lu
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mingyang Chen
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Nengming Lin
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China
| | - Huidi Jiang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| | - Yuqing Wang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China.
| |
Collapse
|
118
|
Lee YH, Yoon AR, Yun CO, Chung KC. Dual-specificity kinase DYRK3 phosphorylates p62 at the Thr-269 residue and promotes melanoma progression. J Biol Chem 2024; 300:107206. [PMID: 38519031 PMCID: PMC11021969 DOI: 10.1016/j.jbc.2024.107206] [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: 11/28/2023] [Revised: 03/03/2024] [Accepted: 03/12/2024] [Indexed: 03/24/2024] Open
Abstract
Melanoma is a type of skin cancer that originates in melanin-producing melanocytes. It is considered a multifactorial disease caused by both genetic and environmental factors, such as UV radiation. Dual-specificity tyrosine-phosphorylation-regulated kinase (DYRK) phosphorylates many substrates involved in signaling pathways, cell survival, cell cycle control, differentiation, and neuronal development. However, little is known about the cellular function of DYRK3, one of the five members of the DYRK family. Interestingly, it was observed that the expression of DYRK3, as well as p62 (a multifunctional signaling protein), is highly enhanced in most melanoma cell lines. This study aimed to investigate whether DYRK3 interacts with p62, and how this affects melanoma progression, particularly in melanoma cell lines. We found that DYRK3 directly phosphorylates p62 at the Ser-207 and Thr-269 residue. Phosphorylation at Thr-269 of p62 by DYRK3 increased the interaction of p62 with tumor necrosis factor receptor-associated factor 6 (TRAF6), an already known activator of mammalian target of rapamycin complex 1 (mTORC1) in the mTOR-involved signaling pathways. Moreover, the phosphorylation of p62 at Thr-269 promoted the activation of mTORC1. We also found that DYRK3-mediated phosphorylation of p62 at Thr-269 enhanced the growth of melanoma cell lines and melanoma progression. Conversely, DYRK3 knockdown or blockade of p62-T269 phosphorylation inhibited melanoma growth, colony formation, and cell migration. In conclusion, we demonstrated that DYRK3 phosphorylates p62, positively modulating the p62-TRAF6-mTORC1 pathway in melanoma cells. This finding suggests that DYRK3 suppression may be a novel therapy for preventing melanoma progression by regulating the mTORC1 pathway.
Collapse
Affiliation(s)
- Ye Hyung Lee
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.
| |
Collapse
|
119
|
Barrow ER, Valionyte E, Baxter CR, Yang Y, Herath S, O'Connell WA, Lopatecka J, Strachan A, Woznica W, Stephenson HN, Fejer G, Sharma V, Lu B, Luo S. Discovery of SQSTM1/p62-dependent P-bodies that regulate the NLRP3 inflammasome. Cell Rep 2024; 43:113935. [PMID: 38460129 DOI: 10.1016/j.celrep.2024.113935] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/22/2024] [Accepted: 02/22/2024] [Indexed: 03/11/2024] Open
Abstract
Autophagy and ribonucleoprotein granules, such as P-bodies (PBs) and stress granules, represent vital stress responses to maintain cellular homeostasis. SQSTM1/p62 phase-separated droplets are known to play critical roles in selective autophagy; however, it is unknown whether p62 can exist as another form in addition to its autophagic droplets. Here, we found that, under stress conditions, including proteotoxicity, endotoxicity, and oxidation, autophagic p62 droplets are transformed to a type of enlarged PBs, termed p62-dependent P-bodies (pd-PBs). p62 phase separation is essential for the nucleation of pd-PBs. Mechanistically, pd-PBs are triggered by enhanced p62 droplet formation upon stress stimulation through the interactions between p62 and DDX6, a DEAD-box ATPase. Functionally, pd-PBs recruit the NLRP3 inflammasome adaptor ASC to assemble the NLRP3 inflammasome and induce inflammation-associated cytotoxicity. Our study shows that p62 droplet-to-PB transformation acts as a stress response to activate the NLRP3 inflammasome process, suggesting that persistent pd-PBs lead to NLRP3-dependent inflammation toxicity.
Collapse
Affiliation(s)
- Elizabeth R Barrow
- Peninsula Medical School, Faculty of Health, University of Plymouth, Research Way, PL6 8BU Plymouth, UK
| | - Evelina Valionyte
- Peninsula Medical School, Faculty of Health, University of Plymouth, Research Way, PL6 8BU Plymouth, UK
| | - Chris R Baxter
- Peninsula Medical School, Faculty of Health, University of Plymouth, Research Way, PL6 8BU Plymouth, UK
| | - Yi Yang
- Peninsula Medical School, Faculty of Health, University of Plymouth, Research Way, PL6 8BU Plymouth, UK
| | - Sharon Herath
- Peninsula Medical School, Faculty of Health, University of Plymouth, Research Way, PL6 8BU Plymouth, UK
| | - William A O'Connell
- Peninsula Medical School, Faculty of Health, University of Plymouth, Research Way, PL6 8BU Plymouth, UK
| | - Justyna Lopatecka
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, PL4 8AA Plymouth, UK
| | - Alexander Strachan
- Plymouth Electron Microscopy Centre, University of Plymouth, Drake Circus, PL4 8AA Plymouth, UK
| | - Waldemar Woznica
- Peninsula Medical School, Faculty of Health, University of Plymouth, Research Way, PL6 8BU Plymouth, UK
| | - Holly N Stephenson
- Peninsula Medical School, Faculty of Health, University of Plymouth, Research Way, PL6 8BU Plymouth, UK
| | - Gyorgy Fejer
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, PL4 8AA Plymouth, UK
| | - Vikram Sharma
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, PL4 8AA Plymouth, UK
| | - Boxun Lu
- State Key Laboratory of Medical Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Shouqing Luo
- Peninsula Medical School, Faculty of Health, University of Plymouth, Research Way, PL6 8BU Plymouth, UK.
| |
Collapse
|
120
|
Wu M, Fan Y, Li L, Yuan J. Bi-directional regulation of type I interferon signaling by heme oxygenase-1. iScience 2024; 27:109185. [PMID: 38420586 PMCID: PMC10901085 DOI: 10.1016/j.isci.2024.109185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/23/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Moderate activation of IFN-I contributes to the body's immune response, but its abnormal expression, stimulated by oxidative stress or other factors causes pathological damage. Heme oxygenase-1 (HO-1), induced by stress stimuli in the body, exerts a central role in cellular protection. Here we showed that HO-1 could promote IFN-1 under Spring Viremia of Carp virus (SVCV) infection and concomitantly attenuate the replication of SVCV. Further characterization of truncated mutants of HO-1 confirmed that intact HO-1 was essential for its antiviral function via IFN-I. Importantly, HO-1 inhibited the IFN-I signal by degrading the IRF3/7 through the autophagy pathway when it was triggered by H2O2 treatment. The iron ion-binding site (His28) was critical for HO-1 to degrade IRF3/7. HO-1 degradation of IRF3/7 is conserved in fish and mammals. Collectively, HO-1 regulates IFN-I positively under viral infection and negatively under oxidative stress, elucidating a mechanism by which HO-1 regulates IFN-I signaling in bi-directions.
Collapse
Affiliation(s)
- Miaomiao Wu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, People’s Republic of China
- Hubei Engineering Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, People’s Republic of China
| | - Yihui Fan
- Hubei Engineering Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, People’s Republic of China
- National Aquatic Animal Diseases Para-reference laboratory (HZAU), Wuhan 430070, People’s Republic of China
| | - Lijuan Li
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, People’s Republic of China
- Hubei Engineering Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, People’s Republic of China
- National Aquatic Animal Diseases Para-reference laboratory (HZAU), Wuhan 430070, People’s Republic of China
| | - Junfa Yuan
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, People’s Republic of China
- Hubei Engineering Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, People’s Republic of China
- National Aquatic Animal Diseases Para-reference laboratory (HZAU), Wuhan 430070, People’s Republic of China
| |
Collapse
|
121
|
Wei Y, Gu Y, Zhou Z, Wu C, Liu Y, Sun H. TRIM21 Promotes Oxidative Stress and Ferroptosis through the SQSTM1-NRF2-KEAP1 Axis to Increase the Titers of H5N1 Highly Pathogenic Avian Influenza Virus. Int J Mol Sci 2024; 25:3315. [PMID: 38542289 PMCID: PMC10970474 DOI: 10.3390/ijms25063315] [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: 01/18/2024] [Revised: 02/26/2024] [Accepted: 03/03/2024] [Indexed: 04/04/2024] Open
Abstract
Tripartite motif-containing protein 21 (TRIM21) is involved in signal transduction and antiviral responses through the ubiquitination of protein targets. TRIM21 was reported to be related to the imbalance of host cell homeostasis caused by viral infection. Our studies indicated that H5N1 highly pathogenic avian influenza virus (HPAIV) infection up-regulated TRIM21 expression in A549 cells. Western blot and qPCR results showed that knockdown of TRIM21 alleviated oxidative stress and ferroptosis induced by H5N1 HPAIV and promoted the activation of antioxidant pathways. Co-IP results showed that TRIM21 promoted oxidative stress and ferroptosis by regulating the SQSTM1-NRF2-KEAP1 axis by increasing SQSTM1 K63-linked polyubiquitination under the condition of HPAIV infection. In addition, TRIM21 attenuated the inhibitory effect of antioxidant NAC on HPAIV titers and enhanced the promoting effect of ferroptosis agonist Erastin on HPAIV titers. Our findings provide new insight into the role of TRIM21 in oxidative stress and ferroptosis induced by viral infection.
Collapse
Affiliation(s)
- Yifan Wei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.W.); (Y.G.); (Z.Z.); (C.W.); (Y.L.)
- Key Laboratory of Zoonosis Control and Prevention of Guangdong Province, South China Agricultural University, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou 510642, China
| | - Yongxia Gu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.W.); (Y.G.); (Z.Z.); (C.W.); (Y.L.)
- Key Laboratory of Zoonosis Control and Prevention of Guangdong Province, South China Agricultural University, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou 510642, China
| | - Ziwei Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.W.); (Y.G.); (Z.Z.); (C.W.); (Y.L.)
- Key Laboratory of Zoonosis Control and Prevention of Guangdong Province, South China Agricultural University, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou 510642, China
| | - Changrong Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.W.); (Y.G.); (Z.Z.); (C.W.); (Y.L.)
- Key Laboratory of Zoonosis Control and Prevention of Guangdong Province, South China Agricultural University, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou 510642, China
| | - Yanwei Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.W.); (Y.G.); (Z.Z.); (C.W.); (Y.L.)
- Key Laboratory of Zoonosis Control and Prevention of Guangdong Province, South China Agricultural University, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou 510642, China
| | - Hailiang Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Y.W.); (Y.G.); (Z.Z.); (C.W.); (Y.L.)
- Key Laboratory of Zoonosis Control and Prevention of Guangdong Province, South China Agricultural University, Guangzhou 510642, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
122
|
Jiang Y, Yu Y, Pan Z, Glandorff C, Sun M. Ferroptosis: a new hunter of hepatocellular carcinoma. Cell Death Discov 2024; 10:136. [PMID: 38480712 PMCID: PMC10937674 DOI: 10.1038/s41420-024-01863-1] [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: 11/07/2023] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 03/17/2024] Open
Abstract
Ferroptosis is an iron ion-dependent, regulatory cell death modality driven by intracellular lipid peroxidation that plays a key role in the development of HCC. Studies have shown that various clinical agents (e.g., sorafenib) have ferroptosis inducer-like effects and can exert therapeutic effects by modulating different key factors in the ferroptosis pathway. This implies that targeting tumor cell ferroptosis may be a very promising strategy for tumor therapy. In this paper, we summarize the prerequisites and defense systems for the occurrence of ferroptosis and the regulatory targets of drug-mediated ferroptosis action in HCC, the differences and connections between ferroptosis and other programmed cell deaths. We aim to summarize the theoretical basis, classical inducers of ferroptosis and research progress of ferroptosis in HCC cells, clued to the treatment of HCC by regulating ferroptosis network. Further investigation of the specific mechanisms of ferroptosis and the development of hepatocellular carcinoma and interventions at different stages of hepatocellular carcinoma will help us to deepen our understanding of hepatocellular carcinoma, with a view to providing new and more precise preventive as well as therapeutic measures for patients.
Collapse
Affiliation(s)
- Yulang Jiang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yongxin Yu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ziyang Pan
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Christian Glandorff
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- University Clinic of Hamburg at the HanseMerkur Center of TCM, Hamburg, Germany
| | - Mingyu Sun
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
123
|
Scalise S, Zannino C, Lucchino V, Lo Conte M, Abbonante V, Benedetto GL, Scalise M, Gambardella A, Parrotta EI, Cuda G. Ascorbic acid mitigates the impact of oxidative stress in a human model of febrile seizure and mesial temporal lobe epilepsy. Sci Rep 2024; 14:5941. [PMID: 38467734 PMCID: PMC10928078 DOI: 10.1038/s41598-024-56680-4] [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: 11/24/2023] [Accepted: 03/09/2024] [Indexed: 03/13/2024] Open
Abstract
Prolonged febrile seizures (FS) in children are linked to the development of temporal lobe epilepsy (MTLE). The association between these two pathologies may be ascribed to the long-term effects that FS exert on neural stem cells, negatively affecting the generation of new neurons. Among the insults associated with FS, oxidative stress is noteworthy. Here, we investigated the consequences of exposure to hydrogen peroxide (H2O2) in an induced pluripotent stem cell-derived neural stem cells (iNSCs) model of a patient affected by FS and MTLE. In our study, we compare the findings from the MTLE patient with those derived from iNSCs of a sibling exhibiting a milder phenotype defined only by FS, as well as a healthy individual. In response to H2O2 treatment, iNSCs derived from MTLE patients demonstrated an elevated production of reactive oxygen species and increased apoptosis, despite the higher expression levels of antioxidant genes and proteins compared to other cell lines analysed. Among the potential causative mechanisms of enhanced vulnerability of MTLE patient iNSCs to oxidative stress, we found that these cells express low levels of the heat shock protein HSPB1 and of the autophagy adaptor SQSTM1/p62. Pre-treatment of diseased iNSCs with the antioxidant molecule ascorbic acid restored HSBP1 and p62 expression and simultaneously reduced the levels of ROS and apoptosis. Our findings suggest the potential for rescuing the impaired oxidative stress response in diseased iNSCs through antioxidant treatment, offering a promising mechanism to prevent FS degeneration in MTLE.
Collapse
Affiliation(s)
- Stefania Scalise
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Clara Zannino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Valeria Lucchino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Michela Lo Conte
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Vittorio Abbonante
- Department of Health Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Giorgia Lucia Benedetto
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Antonio Gambardella
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Elvira Immacolata Parrotta
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy.
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| |
Collapse
|
124
|
Endo A, Fukushima T, Takahashi C, Tsuchiya H, Ohtake F, Ono S, Ly T, Yoshida Y, Tanaka K, Saeki Y, Komada M. USP8 prevents aberrant NF-κB and Nrf2 activation by counteracting ubiquitin signals from endosomes. J Cell Biol 2024; 223:e202306013. [PMID: 38180476 PMCID: PMC10783432 DOI: 10.1083/jcb.202306013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/26/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
K63-linked ubiquitin chains attached to plasma membrane proteins serve as tags for endocytosis and endosome-to-lysosome sorting. USP8 is an essential deubiquitinase for the maintenance of endosomal functions. Prolonged depletion of USP8 leads to cell death, but the major effects on cellular signaling pathways are poorly understood. Here, we show that USP8 depletion causes aberrant accumulation of K63-linked ubiquitin chains on endosomes and induces immune and stress responses. Upon USP8 depletion, two different decoders for K63-linked ubiquitin chains, TAB2/3 and p62, were recruited to endosomes and activated the TAK1-NF-κB and Keap1-Nrf2 pathways, respectively. Oxidative stress, an environmental stimulus that potentially suppresses USP8 activity, induced accumulation of K63-linked ubiquitin chains on endosomes, recruitment of TAB2, and expression of the inflammatory cytokine. The results demonstrate that USP8 is a gatekeeper of misdirected ubiquitin signals and inhibits immune and stress response pathways by removing K63-linked ubiquitin chains from endosomes.
Collapse
Affiliation(s)
- Akinori Endo
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Toshiaki Fukushima
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Chikage Takahashi
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hikaru Tsuchiya
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Fumiaki Ohtake
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
| | - Sayaka Ono
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tony Ly
- Molecular Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Yukiko Yoshida
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Keiji Tanaka
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yasushi Saeki
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Division of Protein Metabolism, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masayuki Komada
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
125
|
Chen F, Wang Q, Xiao M, Lou D, Wufur R, Hu S, Zhang Z, Wang Y, Zhang Y. A novel crosstalk between Nrf2 and Smad2/3 bridged by two nuanced Keap1 isoforms with their divergent effects on these distinct family transcription factors. Free Radic Biol Med 2024; 213:190-207. [PMID: 38242246 DOI: 10.1016/j.freeradbiomed.2024.01.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
The Keap1-Nrf2 signalling to transcriptionally regulate antioxidant response element (ARE)-driven target genes has been accepted as key redox-sensitive pathway governing a vast variety of cellular stresses during healthy survival and disease development. Herein, we identified two nuanced isoforms α and β of Keap1 in HepG2 cells, arising from its first and another in-frame translation starting codons, respectively. In identifying those differential expression genes monitored by Keap1α and/or Keap1β, an unusual interaction of Keap1 with Smad2/3 was discovered by parsing transcriptome sequencing, Keap1-interacting protein profiling and relevant immunoprecipitation data. Further examination validated that Smad2/3 enable physical interaction with Keap1, as well as its isoforms α and β, by both EDGETSD and DLG motifs in the linker regions between their MH1 and MH2 domains, such that the stability of Smad2/3 and transcriptional activity are enhanced with their prolonged half-lives and relevant signalling responses from the cytoplasmic to nuclear compartments. The activation of Smad2/3 by Keap1, Keap1α or Keap1β was much likely contributable to a coordinative or another competitive effect of Nrf2, particularly in distinct Keap1-based cellular responses to its cognate growth factor (i.e. TGF-β1) or redox stress (e.g. stimulated by tBHQ and DTT). Overall, this discovery presents a novel functional bridge crossing the Keap1-Nrf2 redox signalling and the TGF-β1-Smad2/3 pathways so as to coordinately regulate the healthy growth and development.
Collapse
Affiliation(s)
- Feilong Chen
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402262, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Qing Wang
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402262, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Mei Xiao
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Deshuai Lou
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, China
| | - Reziyamu Wufur
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402262, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Shaofan Hu
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Zhengwen Zhang
- Laboratory of Neuroscience, Institute of Cognitive Neuroscience and School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, England, United Kingdom
| | - Yeqi Wang
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Yiguo Zhang
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402262, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China.
| |
Collapse
|
126
|
Chen L, Qiu H, Chen Q, Xiang P, Lei J, Zhang J, Lu Y, Wang X, Wu S, Yu C, Ma L. N-acetylneuraminic acid modulates SQSTM1/p62 sialyation-mediated ubiquitination degradation contributing to vascular endothelium dysfunction in experimental atherosclerosis mice. IUBMB Life 2024; 76:161-178. [PMID: 37818680 DOI: 10.1002/iub.2788] [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: 06/23/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023]
Abstract
Sialic acid (SIA) has been reported to be a risk factor for atherosclerosis (AS) due to its high plasma levels in such patients. However, the effect of increasing SIA in circulation on endothelial function during AS progression remains unclear. In the present study, ApoE-/- mice and endothelial cells line (HUVEC cells) were applied to investigate the effect of SIA on AS progression and its potential molecular mechanism. In vivo, mice were injected intraperitoneally with Neu5Ac (main form of SIA) to keep high-level SIA in circulation. ORO, H&E, and Masson staining were applied to detect the plaque progression. In vitro, HUVECs were treated with Neu5Ac at different times, CCK-8, RT-PCR, western blot, and immunoprecipitation methods were used to analyze its effects on endothelial function and the potential involved mechanism. Results from the present study showed that high plasma levels of Neu5Ac in ApoE-/- mice could aggravate the plaque areas as well as increase necrotic core areas and collagen fiber contents. Remarkably, Neu5Ac levels in circulation displayed a positive correlation with AS plaque areas. Furthermore, results from HUVECs showed that Neu5Ac inhibited cells viability in a time/dose-dependent manner, by then induced the activation of inflammation makers such as ICAM-1 and IL-1β. Mechanism study showed that the activation of excessive autophagy medicated by SQSTM1/p62 displayed an important role in endothelium inflammatory injury. Neu5Ac could modify SQSTM1/p62 as a sialylation protein, and then increase its level with ubiquitin binding, further inducing ubiquitination degradation and being involved in the excessive autophagy pathway. Inhibition of sialylation by P-3Fax-Neu5Ac, a sialyltransferase inhibitor, reduced the binding of SQSTM1/p62 to ubiquitin. Together, these findings indicated that Neu5Ac increased SQSTM1/p62-ubiquitin binding through sialylation modification, thereby inducing excessive autophagy and subsequent endothelial injury. Inhibition of SQSTM1/p62 sialylation might be a potential strategy for preventing such disease with high levels of Neu5Ac in circulation.
Collapse
Affiliation(s)
- Le Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Hongmei Qiu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Qingqiu Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Peng Xiang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Jin Lei
- Xi'an No.1 Hospital, The First Affiliated Hospital of Northwest University, Xi'an, China
| | - Jun Zhang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Yining Lu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Xianmin Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Shengde Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Limei Ma
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| |
Collapse
|
127
|
Gurubaran IS. Mitochondrial damage and clearance in retinal pigment epithelial cells. Acta Ophthalmol 2024; 102 Suppl 282:3-53. [PMID: 38467968 DOI: 10.1111/aos.16661] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 03/13/2024]
Abstract
Age-related macular degeneration (AMD) is a devastating eye disease that causes permanent vision loss in the central part of the retina, known as the macula. Patients with such severe visual loss face a reduced quality of life and are at a 1.5 times greater risk of death compared to the general population. Currently, there is no cure for or effective treatment for dry AMD. There are several mechanisms thought to underlie the disease, for example, ageing-associated chronic oxidative stress, mitochondrial damage, harmful protein aggregation and inflammation. As a way of gaining a better understanding of the molecular mechanisms behind AMD and thus developing new therapies, we have created a peroxisome proliferator-activated receptor gamma coactivator 1-alpha and nuclear factor erythroid 2-related factor 2 (PGC1α/NFE2L2) double-knockout (dKO) mouse model that mimics many of the clinical features of dry AMD, including elevated levels of oxidative stress markers, damaged mitochondria, accumulating lysosomal lipofuscin and extracellular drusen-like structures in retinal pigment epithelial cells (RPE). In addition, a human RPE cell-based model was established to examine the impact of non-functional intracellular clearance systems on inflammasome activation. In this study, we found that there was a disturbance in the autolysosomal machinery responsible for clearing mitochondria in the RPE cells of one-year-old PGC1α/NFE2L2-deficient mice. The confocal immunohistochemical analysis revealed an increase in autophagosome marker microtubule-associated proteins 1A/1B light chain 3B (LC3B) as well as multiple mitophagy markers such as PTE-induced putative kinase 1 (PINK1) and E3 ubiquitin ligase (PARKIN), along with signs of damaged mitochondria. However, no increase in autolysosome formation was detected, nor was there a colocalization of the lysosomal marker LAMP2 or the mitochondrial marker, ATP synthase β. There was an upregulation of late autolysosomal fusion Ras-related protein (Rab7) in the perinuclear space of RPE cells, together with autofluorescent aggregates. Additionally, we observed an increase in the numbers of Toll-like receptors 3 and 9, while those of NOD-like receptor 3 were decreased in PGC1α/NFE2L2 dKO retinal specimens compared to wild-type animals. There was a trend towards increased complement component C5a and increased involvement of the serine protease enzyme, thrombin, in enhancing the terminal pathway producing C5a, independent of C3. The levels of primary acute phase C-reactive protein and receptor for advanced glycation end products were also increased in the PGC1α/NFE2L2 dKO retina. Furthermore, selective proteasome inhibition with epoxomicin promoted both nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondrial-mediated oxidative stress, leading to the release of mitochondrial DNA to the cytosol, resulting in potassium efflux-dependent activation of the absent in melanoma 2 (AIM2) inflammasome and the subsequent secretion of interleukin-1β in ARPE-19 cells. In conclusion, the data suggest that there is at least a relative decrease in mitophagy, increases in the amounts of C5 and thrombin and decreased C3 levels in this dry AMD-like model. Moreover, selective proteasome inhibition evoked mitochondrial damage and AIM2 inflammasome activation in ARPE-19 cells.
Collapse
Affiliation(s)
- Iswariyaraja Sridevi Gurubaran
- Department of Medicine, Clinical Medicine Unit, University of Eastern Finland Institute of Clinical Medicine, Kuopio, Northern Savonia, Finland
| |
Collapse
|
128
|
Xiang Y, Song X, Long D. Ferroptosis regulation through Nrf2 and implications for neurodegenerative diseases. Arch Toxicol 2024; 98:579-615. [PMID: 38265475 PMCID: PMC10861688 DOI: 10.1007/s00204-023-03660-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/07/2023] [Indexed: 01/25/2024]
Abstract
This article provides an overview of the background knowledge of ferroptosis in the nervous system, as well as the key role of nuclear factor E2-related factor 2 (Nrf2) in regulating ferroptosis. The article takes Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) as the starting point to explore the close association between Nrf2 and ferroptosis, which is of clear and significant importance for understanding the mechanism of neurodegenerative diseases (NDs) based on oxidative stress (OS). Accumulating evidence links ferroptosis to the pathogenesis of NDs. As the disease progresses, damage to the antioxidant system, excessive OS, and altered Nrf2 expression levels, especially the inhibition of ferroptosis by lipid peroxidation inhibitors and adaptive enhancement of Nrf2 signaling, demonstrate the potential clinical significance of Nrf2 in detecting and identifying ferroptosis, as well as targeted therapy for neuronal loss and mitochondrial dysfunction. These findings provide new insights and possibilities for the treatment and prevention of NDs.
Collapse
Affiliation(s)
- Yao Xiang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China
| | - Xiaohua Song
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China
| | - Dingxin Long
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China.
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China.
| |
Collapse
|
129
|
Baumel-Alterzon S, Katz LS, Lambertini L, Tse I, Heidery F, Garcia-Ocaña A, Scott DK. NRF2 is required for neonatal mouse beta cell growth by maintaining redox balance and promoting mitochondrial biogenesis and function. Diabetologia 2024; 67:547-560. [PMID: 38206362 PMCID: PMC11521447 DOI: 10.1007/s00125-023-06071-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/13/2023] [Indexed: 01/12/2024]
Abstract
AIMS/HYPOTHESIS All forms of diabetes result from insufficient functional beta cell mass. Due to the relatively limited expression of several antioxidant enzymes, beta cells are highly vulnerable to pathological levels of reactive oxygen species (ROS), which can lead to the reduction of functional beta cell mass. During early postnatal ages, both human and rodent beta cells go through a burst of proliferation that quickly declines with age. The exact mechanisms that account for neonatal beta cell proliferation are understudied but mitochondrial release of moderated ROS levels has been suggested as one of the main drivers. We previously showed that, apart from its conventional role in protecting beta cells from oxidative stress, the nuclear factor erythroid 2-related factor 2 (NRF2) is also essential for beta cell proliferation. We therefore hypothesised that NRF2, which is activated by ROS, plays an essential role in beta cell proliferation at early postnatal ages. METHODS Beta cell NRF2 levels and beta cell proliferation were measured in pancreatic sections from non-diabetic human cadaveric donors at different postnatal ages, childhood and adulthood. Pancreatic sections from 1-, 7-, 14- and 28-day-old beta cell-specific Nrf2 (also known as Nfe2l2)-knockout mice (βNrf2KO) or control (Nrf2lox/lox) mice were assessed for beta cell NRF2 levels, beta cell proliferation, beta cell oxidative stress, beta cell death, nuclear beta cell pancreatic duodenal homeobox protein 1 (PDX1) levels and beta cell mass. Seven-day-old βNrf2KO and Nrf2lox/lox mice were injected daily with N-acetylcysteine (NAC) or saline (154 mmol/l NaCl) to explore the potential contribution of oxidative stress to the phenotypes seen in βNrf2KO mice at early postnatal ages. RNA-seq was performed on 7-day-old βNrf2KO and Nrf2lox/lox mice to investigate the mechanisms by which NRF2 stimulates beta cell proliferation at early postnatal ages. Mitochondrial biogenesis and function were determined using dispersed islets from 7-day-old βNrf2KO and Nrf2lox/lox mice by measuring MitoTracker intensity, mtDNA/gDNA ratio and ATP/ADP ratio. To study the effect of neonatal beta cell-specific Nrf2 deletion on glucose homeostasis in adulthood, blood glucose, plasma insulin and insulin secretion were determined and a GTT was performed on 3-month-old βNrf2KO and Nrf2lox/lox mice fed on regular diet (RD) or high-fat diet (HFD). RESULTS The expression of the master antioxidant regulator NRF2 was increased at early postnatal ages in both human (1 day to 19 months old, 31%) and mouse (7 days old, 57%) beta cells, and gradually declined with age (8% in adult humans, 3.77% in adult mice). A significant correlation (R2=0.568; p=0.001) was found between beta cell proliferation and NRF2 levels in human beta cells. Seven-day-old βNrf2KO mice showed reduced beta cell proliferation (by 65%), beta cell nuclear PDX1 levels (by 23%) and beta cell mass (by 67%), and increased beta cell oxidative stress (threefold) and beta cell death compared with Nrf2lox/lox control mice. NAC injections increased beta cell proliferation in 7-day-old βNrf2KO mice (3.4-fold) compared with saline-injected βNrf2KO mice. Interestingly, RNA-seq of islets isolated from 7-day-old βNrf2KO mice revealed reduced expression of mitochondrial RNA genes and genes involved in the electron transport chain. Islets isolated from 7-day old βNrf2KO mice presented reduced MitoTracker intensity (by 47%), mtDNA/gDNA ratio (by 75%) and ATP/ADP ratio (by 68%) compared with islets from Nrf2lox/lox littermates. Lastly, HFD-fed 3-month-old βNrf2KO male mice displayed a significant reduction in beta cell mass (by 35%), a mild increase in non-fasting blood glucose (1.2-fold), decreased plasma insulin (by 14%), and reduced glucose tolerance (1.3-fold) compared with HFD-fed Nrf2lox/lox mice. CONCLUSIONS/INTERPRETATION Our study highlights NRF2 as an essential transcription factor for maintaining neonatal redox balance, mitochondrial biogenesis and function and beta cell growth, and for preserving functional beta cell mass in adulthood under metabolic stress. DATA AVAILABILITY Sequencing data are available in the NCBI Gene Expression Omnibus, accession number GSE242718 ( https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE242718 ).
Collapse
Affiliation(s)
- Sharon Baumel-Alterzon
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Liora S Katz
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luca Lambertini
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isabelle Tse
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fatema Heidery
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adolfo Garcia-Ocaña
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute at City of Hope, Duarte, CA, USA
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
130
|
Fan M, Huo S, Guo Y, Wang R, Hao W, Zhang Z, Wang L, Zhao Y. UDP-glucose dehydrogenase supports autophagy-deficient PDAC growth via increasing hyaluronic acid biosynthesis. Cell Rep 2024; 43:113808. [PMID: 38367236 DOI: 10.1016/j.celrep.2024.113808] [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: 09/19/2023] [Revised: 01/04/2024] [Accepted: 02/01/2024] [Indexed: 02/19/2024] Open
Abstract
Autophagy is an essential degradation and recycling process that maintains cellular homeostasis during stress or nutrient deprivation. However, certain types of tumors such as pancreatic cancers can circumvent autophagy inhibition to sustain growth. The mechanism that autophagy-deficient pancreatic ductal adenocarcinoma (PDAC) uses to grow under nutrient deprivation is poorly understood. Our data show that nutrient deprivation in PDAC results in UDP-glucose dehydrogenase (UGDH) degradation, which is dependent on autophagic cargo receptor sequestosome 1 (p62). Moreover, we demonstrate that accumulated UGDH is indispensable for autophagy-deficient PDAC cells proliferation by promoting hyaluronic acid (HA) synthesis upon energy deprivation. Using an orthotopic mouse model of PDAC, we find that inhibition of HA synthesis by targeting UGDH in PDAC reduces tumor weight. Thus, the combined inhibition of HA and autophagy might be an attractive strategy for PDAC treatment.
Collapse
Affiliation(s)
- Minghe Fan
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Sihan Huo
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Yuyao Guo
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Ruoxuan Wang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Wenqin Hao
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Ziyang Zhang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Lina Wang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Ying Zhao
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China; Peking University Cancer Hospital and Institute, Beijing 100142, China.
| |
Collapse
|
131
|
Kurusu R, Morishita H, Komatsu M. p62 bodies: cytosolic zoning by phase separation. J Biochem 2024; 175:141-146. [PMID: 37948628 DOI: 10.1093/jb/mvad089] [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: 09/07/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Cellular zoning or partitioning is critical in preventing macromolecules from random diffusion and orchestrating the spatiotemporal dynamics of biochemical reactions. Along with membranous organelles, membraneless organelles contribute to the precise regulation of biochemical reactions inside cells. In response to environmental cues, membraneless organelles rapidly form through liquid-liquid phase separation, sequester certain proteins and RNAs, mediate specific reactions and dissociate. Among membraneless organelles, ubiquitin-positive condensates, namely, p62 bodies, maintain cellular homeostasis through selective autophagy of themselves to contribute to intracellular quality control. p62 bodies also activate the anti-oxidative stress response regulated by the KEAP1-NRF2 system. In this review, we present an overview of recent advancements in cellular and molecular biology related to p62 bodies, highlighting their dynamic nature and functions.
Collapse
Affiliation(s)
- Reo Kurusu
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hideaki Morishita
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
132
|
Gęgotek A, Skrzydlewska E. Lipid peroxidation products' role in autophagy regulation. Free Radic Biol Med 2024; 212:375-383. [PMID: 38182071 DOI: 10.1016/j.freeradbiomed.2024.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 01/07/2024]
Abstract
Autophagy, which is responsible for removing damaged molecules, prevents their accumulation in cells, thus maintaining intracellular homeostasis. It is also responsible for removing the effects of oxidative stress, so its activation takes place during increased reactive oxygen species (ROS) generation and lipid peroxidation. Therefore, the aim of this review was to summarize all the available knowledge about the effect of protein modifications by lipid peroxidation products on autophagy activation and the impact of this interaction on the functioning of cells. This review shows that reactive aldehydes (including 4-hydroxynonenal and malondialdehyde), either directly or by the formation of adducts with autophagic proteins, can activate or prevent autophagy, depending on their concentration. This effect relates not only to the initial stages of autophagy, when 4-hydroxynonenal and malondialdehyde affect the levels of proteins involved in autophagy initiation and phagophore formation, but also to the final stage, degradation, when reactive aldehydes, by binding to the active center of cathepsins, inactivate their proteolytic functions. Moreover, this review also shows how little research exists on analyzing the impact of lipid peroxidation products and their protein adducts on autophagy. Such knowledge could be used in the therapy of diseases related to autophagy disorders.
Collapse
Affiliation(s)
- Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-069, Bialystok, Poland.
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-069, Bialystok, Poland
| |
Collapse
|
133
|
Wang Y, Lyu L, Vu T, McCarty N. WITHDRAWN: TRIM44 promotes autophagy through SQSTM1 oligomerization in the response to oxidative stress induced by Arsenic Trioxide in cancer cells. RESEARCH SQUARE 2024:rs.3.rs-3951960. [PMID: 38464079 PMCID: PMC10925436 DOI: 10.21203/rs.3.rs-3951960/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The authors have requested that this preprint be removed from Research Square.
Collapse
|
134
|
Li X, Wang S, Zhang M, Li M. The SLC38A9-mTOR axis is involved in autophagy in the juvenile yellow catfish (Pelteobagrus fulvidraco) under ammonia stress. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 343:123211. [PMID: 38142034 DOI: 10.1016/j.envpol.2023.123211] [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: 06/16/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 12/25/2023]
Abstract
The primary objective of this study was to examine the effect of acute ammonia stress on hepatic physiological alterations in yellow catfish by performing a comprehensive analysis of the metabolome and transcriptome. The present study showed that ammonia stress led to liver metabolic disruption, functional incapacitation, and oxidative damage. Transcriptomic and metabolomic analyses revealed transcriptional and metabolic differences in the liver of yellow catfish under control and high ammonia stress conditions. After 96 h of acute exposure to ammonia, the mRNA levels of 596 liver genes were upregulated, whereas those of 603 genes were downregulated. Enrichment analysis of the differentially expressed genes identified multiple signalling pathways associated with autophagy, including the endocytosis, autophagy-animal, and mammalian target of rapamycin signalling pathways. A total of 186 upregulated and 117 downregulated metabolites, primarily associated with amino acid biosynthesis pathways, were identified. Multi-omics integration revealed the solute carrier family 38 member 9 (SLC38A9)-mammalian target of rapamycin axis as a signalling nexus for amino acid-mediated modulation of autophagy flux, and q-PCR was used to assess the expression of autophagy-related genes (LC3a and sqstm1), revealing an initial inhibition followed by the restoration of autophagic flux during ammonia stress. Subsequent utilisation of arginine as a specific SLC38A9 activator during ammonia stress demonstrated that augmented SLC38A9 expression hindered autophagy, exacerbated ammonia toxicity, and caused a physiological decline (total cholesterol, total triglyceride, acid phosphatase, alkaline phosphatase, aspartate aminotransferase, and alanine aminotransferase levels were significantly increased), oxidative stress, and apoptosis. Autophagy activation may be an adaptive mechanism to resist ammonia stress.
Collapse
Affiliation(s)
- Xue Li
- School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Shidong Wang
- School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Muzi Zhang
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Ming Li
- School of Marine Sciences, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
135
|
Lee J, Ou JHJ. HCV-induced autophagy and innate immunity. Front Immunol 2024; 15:1305157. [PMID: 38370419 PMCID: PMC10874285 DOI: 10.3389/fimmu.2024.1305157] [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: 11/14/2023] [Accepted: 01/19/2024] [Indexed: 02/20/2024] Open
Abstract
The interplay between autophagy and host innate immunity has been of great interest. Hepatitis C virus (HCV) impedes signaling pathways initiated by pattern-recognition receptors (PRRs) that recognize pathogens-associated molecular patterns (PAMPs). Autophagy, a cellular catabolic process, delivers damaged organelles and protein aggregates to lysosomes for degradation and recycling. Autophagy is also an innate immune response of cells to trap pathogens in membrane vesicles for removal. However, HCV controls the autophagic pathway and uses autophagic membranes to enhance its replication. Mitophagy, a selective autophagy targeting mitochondria, alters the dynamics and metabolism of mitochondria, which play important roles in host antiviral responses. HCV also alters mitochondrial dynamics and promotes mitophagy to prevent premature cell death and attenuate the interferon (IFN) response. In addition, the dysregulation of the inflammasomal response by HCV leads to IFN resistance and immune tolerance. These immune evasion properties of HCV allow HCV to successfully replicate and persist in its host cells. In this article, we discuss HCV-induced autophagy/mitophagy and its associated immunological responses and provide a review of our current understanding of how these processes are regulated in HCV-infected cells.
Collapse
Affiliation(s)
| | - J.-H. James Ou
- Department of Molecular Microbiology and Immunology, University of Southern California, Keck School of Medicine, Los Angeles, CA, United States
| |
Collapse
|
136
|
Fang H, Shi X, Wan J, Zhong X. Role of sestrins in metabolic and aging-related diseases. Biogerontology 2024; 25:9-22. [PMID: 37516672 DOI: 10.1007/s10522-023-10053-y] [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: 04/29/2023] [Accepted: 07/14/2023] [Indexed: 07/31/2023]
Abstract
Sestrins are a type of highly conserved stress-inducing protein that has antioxidant and mTORC1 inhibitory functions. Metabolic dysfunction and aging are the main risk factors for development of human diseases, such as diabetes, neurodegenerative diseases, and cancer. Sestrins have important roles in regulating glucose and lipid metabolism, anti-tumor functions, and aging by inhibiting the reactive oxygen species and mechanistic target of rapamycin complex 1 pathways. In this review, the structure and biological functions of sestrins are summarized, and how sestrins are activated and contribute to regulation of the downstream signal pathways of metabolic and aging-related diseases are discussed in detail with the goal of providing new ideas and therapeutic targets for the treatment of related diseases.
Collapse
Affiliation(s)
- Huan Fang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, China
| | - Xiaomin Shi
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, China.
| | - Xiaolin Zhong
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, China.
| |
Collapse
|
137
|
Pan M, Hu T, Lyu J, Yin Y, Sun J, Wang Q, Xu L, Hu H, Wang C. CSNK1A1/CK1α suppresses autoimmunity by restraining the CGAS-STING1 signaling. Autophagy 2024; 20:311-328. [PMID: 37723657 PMCID: PMC10813568 DOI: 10.1080/15548627.2023.2256135] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 09/20/2023] Open
Abstract
STING1 (stimulator of interferon response cGAMP interactor 1) is the quintessential protein in the CGAS-STING1 signaling pathway, crucial for the induction of type I IFN (interferon) production and eliciting innate immunity. Nevertheless, the overactivation or sustained activation of STING1 has been closely associated with the onset of autoimmune disorders. Notably, the majority of these disorders manifest as an upregulated expression of type I interferons and IFN-stimulated genes (ISGs). Hence, strict regulation of STING1 activity is paramount to preserve immune homeostasis. Here, we reported that CSNK1A1/CK1α, a serine/threonine protein kinase, was essential to prevent the overactivation of STING1-mediated type I IFN signaling through autophagic degradation of STING1. Mechanistically, CSNK1A1 interacted with STING1 upon the CGAS-STING1 pathway activation and promoted STING1 autophagic degradation by enhancing the phosphorylation of SQSTM1/p62 at serine 351 (serine 349 in human), which was critical for SQSTM1-mediated STING1 autophagic degradation. Consistently, SSTC3, a selective CSNK1A1 agonist, significantly attenuated the response of the CGAS-STING1 signaling by promoting STING1 autophagic degradation. Importantly, pharmacological activation of CSNK1A1 using SSTC3 markedly repressed the systemic autoinflammatory responses in the trex1-/- mouse autoimmune disease model and effectively suppressed the production of IFNs and ISGs in the PBMCs of SLE patients. Taken together, our study reveals a novel regulatory role of CSNK1A1 in the autophagic degradation of STING1 to maintain immune homeostasis. Manipulating CSNK1A1 through SSTC3 might be a potential therapeutic strategy for alleviating STING1-mediated aberrant type I IFNs in autoimmune diseases.Abbreviations: BMDMs: bone marrow-derived macrophages; cGAMP: cyclic GMP-AMP; CGAS: cyclic GMP-AMP synthase; HTDNA: herring testes DNA; IFIT1: interferon induced protein with tetratricopeptide repeats 1; IFNA4: interferon alpha 4; IFNB: interferon beta; IRF3: interferon regulatory factor 3; ISD: interferon stimulatory DNA; ISGs: IFN-stimulated genes; MEFs: mouse embryonic fibroblasts; PBMCs: peripheral blood mononuclear cells; RSAD2: radical S-adenosyl methionine domain containing 2; SLE: systemic lupus erythematosus; STING1: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1.
Collapse
Affiliation(s)
- Mingyu Pan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
- Department of Biomedical Science, City University of Hong Kong, Hong Kong, Hong Kong, China
| | - Tongyu Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jiao Lyu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jing Sun
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lingxiao Xu
- Department of Rheumatology, The affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, Jiangsu, China
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haiyang Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
138
|
Yang S, Li W, Bai X, Di Nunzio G, Fan L, Zhao Y, Ren L, Zhao R, Bian S, Liu M, Wei Y, Zhao D, Wang J. Ginseng-derived nanoparticles alleviate inflammatory bowel disease via the TLR4/MAPK and p62/Nrf2/Keap1 pathways. J Nanobiotechnology 2024; 22:48. [PMID: 38302938 PMCID: PMC10832157 DOI: 10.1186/s12951-024-02313-x] [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: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/03/2024] Open
Abstract
Inflammatory bowel disease (IBD) is closely linked to the homeostasis of the intestinal environment, and exosomes can be used to treat IBD due to their high biocompatibility and ability to be effectively absorbed by the intestinal tract. However, Ginseng-derived nanoparticles (GDNPs) have not been studied in this context and their mechanism of action remains unclear. Here, we investigated GDNPs ability to mediate intercellular communication in a complex inflammatory microenvironment in order to treat IBD. We found that GDNPs scavenge reactive oxygen species from immune cells and intestinal epithelial cells, inhibit the expression of pro-inflammatory factors, promote the proliferation and differentiation of intestinal stem cells, as well as enhancing the diversity of the intestinal flora. GDNPs significantly stabilise the intestinal barrier thereby promoting tissue repair. Overall, we proved that GDNPs can ameliorate inflammation and oxidative stress in vivo and in vitro, acting on the TLR4/MAPK and p62/Keap1/Nrf2 pathways, and exerting an anti-inflammatory and antioxidant effect. GDNPs mitigated IBD in mice by reducing inflammatory factors and improving the intestinal environment. This study offers new evidence of the potential therapeutic effects of GDNPs in the context of IBD, providing the conceptual ground for an alternative therapeutic strategy.
Collapse
Affiliation(s)
- Song Yang
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Wenjing Li
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Xueyuan Bai
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Giada Di Nunzio
- Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Liangliang Fan
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Yueming Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Limei Ren
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Ronghua Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Shuai Bian
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Meichen Liu
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Yuchi Wei
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Daqing Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China
| | - Jiawen Wang
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China.
- Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, 17176, Stockholm, Sweden.
| |
Collapse
|
139
|
Endo M, Tanaka Y, Fukuoka M, Suzuki H, Minami Y. Wnt5a/Ror2 promotes Nrf2-mediated tissue protective function of astrocytes after brain injury. Glia 2024; 72:411-432. [PMID: 37904612 DOI: 10.1002/glia.24483] [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: 05/12/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/01/2023]
Abstract
Astrocytes, a type of glial cells, play critical roles in promoting the protection and repair of damaged tissues after brain injury. Inflammatory cytokines and growth factors can affect gene expression in astrocytes in injured brains, but signaling pathways and transcriptional mechanisms that regulate tissue protective functions of astrocytes are still poorly understood. In this study, we investigated the molecular mechanisms regulating the function of reactive astrocytes induced in mouse models of stab wound (SW) brain injury and collagenase-induced intracerebral hemorrhage (ICH). We show that basic fibroblast growth factor (bFGF), whose expression is up-regulated in mouse brains after SW injury and ICH, acts synergistically with inflammatory cytokines to activate E2F1-mediated transcription of a gene encoding the Ror-family protein Ror2, a receptor for Wnt5a, in cultured astrocytes. We also found that subsequent activation of Wnt5a/Ror2 signaling in astrocytes results in nuclear accumulation of antioxidative transcription factor Nrf2 at least partly by increased expression of p62/Sqstm1, leading to promoted expression of several Nrf2 target genes, including heme oxygenase 1. Finally, we provide evidence demonstrating that enhanced activation of Wnt5a/Ror2 signaling in astrocytes reduces cellular damage caused by hemin, a degradation product of hemoglobin, and promotes repair of the damaged blood brain barrier after brain hemorrhage.
Collapse
Affiliation(s)
- Mitsuharu Endo
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yuki Tanaka
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Mayo Fukuoka
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Hayata Suzuki
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| |
Collapse
|
140
|
Karagiannis D, Wu W, Li A, Hayashi M, Chen X, Yip M, Mangipudy V, Xu X, Sánchez-Rivera FJ, Soto-Feliciano YM, Ye J, Papagiannakopoulos T, Lu C. Metabolic reprogramming by histone deacetylase inhibition preferentially targets NRF2-activated tumors. Cell Rep 2024; 43:113629. [PMID: 38165806 PMCID: PMC10853943 DOI: 10.1016/j.celrep.2023.113629] [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: 12/06/2022] [Revised: 10/27/2023] [Accepted: 12/12/2023] [Indexed: 01/04/2024] Open
Abstract
The interplay between metabolism and chromatin signaling is implicated in cancer progression. However, whether and how metabolic reprogramming in tumors generates chromatin vulnerabilities remain unclear. Lung adenocarcinoma (LUAD) tumors frequently harbor aberrant activation of the NRF2 antioxidant pathway, which drives aggressive and chemo-resistant disease. Using a chromatin-focused CRISPR screen, we report that NRF2 activation sensitizes LUAD cells to genetic and chemical inhibition of class I histone deacetylases (HDACs). This association is observed across cultured cells, mouse models, and patient-derived xenografts. Integrative epigenomic, transcriptomic, and metabolomic analysis demonstrates that HDAC inhibition causes widespread redistribution of H4ac and its reader protein, which transcriptionally downregulates metabolic enzymes. This results in reduced flux into amino acid metabolism and de novo nucleotide synthesis pathways that are preferentially required for the survival of NRF2-active cancer cells. Together, our findings suggest NRF2 activation as a potential biomarker for effective repurposing of HDAC inhibitors to treat solid tumors.
Collapse
Affiliation(s)
- Dimitris Karagiannis
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Warren Wu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Albert Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Makiko Hayashi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Xiao Chen
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michaela Yip
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Vaibhav Mangipudy
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xinjing Xu
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Francisco J Sánchez-Rivera
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Yadira M Soto-Feliciano
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Chao Lu
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
141
|
Li C, Liu Y. Puerarin reduces cell damage from cerebral ischemia-reperfusion by inhibiting ferroptosis. Biochem Biophys Res Commun 2024; 693:149324. [PMID: 38101001 DOI: 10.1016/j.bbrc.2023.149324] [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: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023]
Abstract
This study explores the protective effects of Puerarin, a compound derived from the traditional Chinese herb Pueraria, against cellular damage induced by Oxygen-Glucose Deprivation/Reoxygenation (OGD/R) in PC12 cells. The research focuses on understanding how Puerarin influences the mechanisms of ferroptosis and oxidative stress, key factors in ischemia-reperfusion injury relevant to neurodegenerative diseases. In our in vitro model, we identified the optimal OGD duration to induce significant cell stress and confirmed the non-toxicity of Puerarin up to 100uM. The results reveal that Puerarin substantially mitigates the detrimental effects of OGD/R, including improvements in cell viability, mitochondrial integrity, and reductions in oxidative stress markers like ROS and lipid peroxidation. Notably, Puerarin modulates key proteins in the autophagy process and the Nrf2 pathway, crucial in cellular stress responses. Further, the use of 3-Methyladenine, an autophagy inhibitor, underscores the significance of autophagy in managing OGD/R-induced stress. These findings suggest Puerarin's potential as a therapeutic agent for conditions characterized by ischemic cellular damage, highlighting the need for further clinical exploration.
Collapse
Affiliation(s)
- Changxuan Li
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan, China
| | - Yu Liu
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan, China.
| |
Collapse
|
142
|
Wang Z, Liu N, Yang Y, Tu Z. The novel mechanism facilitating chronic hepatitis B infection: immunometabolism and epigenetic modification reprogramming. Front Immunol 2024; 15:1349867. [PMID: 38288308 PMCID: PMC10822934 DOI: 10.3389/fimmu.2024.1349867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Abstract
Hepatitis B Virus (HBV) infections pose a global public health challenge. Despite extensive research on this disease, the intricate mechanisms underlying persistent HBV infection require further in-depth elucidation. Recent studies have revealed the pivotal roles of immunometabolism and epigenetic reprogramming in chronic HBV infection. Immunometabolism have identified as the process, which link cell metabolic status with innate immunity functions in response to HBV infection, ultimately contributing to the immune system's inability to resolve Chronic Hepatitis B (CHB). Within hepatocytes, HBV replication leads to a stable viral covalently closed circular DNA (cccDNA) minichromosome located in the nucleus, and epigenetic modifications in cccDNA enable persistence of infection. Additionally, the accumulation or depletion of metabolites not only directly affects the function and homeostasis of immune cells but also serves as a substrate for regulating epigenetic modifications, subsequently influencing the expression of antiviral immune genes and facilitating the occurrence of sustained HBV infection. The interaction between immunometabolism and epigenetic modifications has led to a new research field, known as metabolic epigenomics, which may form a mutually reinforcing relationship with CHB. Herein, we review the recent studies on immunometabolism and epigenetic reprogramming in CHB infection and discuss the potential mechanisms of persistent HBV infection. A deeper understanding of these mechanisms will offer novel insights and targets for intervention strategies against chronic HBV infection, thereby providing new hope for the treatment of related diseases.
Collapse
Affiliation(s)
- Zhengmin Wang
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Nan Liu
- Institute of Epigenetic Medicine, First Hospital of Jilin University, Changchun, China
| | - Yang Yang
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhengkun Tu
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
- Institute of Liver Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
143
|
Glorieux C, Enríquez C, González C, Aguirre-Martínez G, Buc Calderon P. The Multifaceted Roles of NRF2 in Cancer: Friend or Foe? Antioxidants (Basel) 2024; 13:70. [PMID: 38247494 PMCID: PMC10812565 DOI: 10.3390/antiox13010070] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Physiological concentrations of reactive oxygen species (ROS) play vital roles in various normal cellular processes, whereas excessive ROS generation is central to disease pathogenesis. The nuclear factor erythroid 2-related factor 2 (NRF2) is a critical transcription factor that regulates the cellular antioxidant systems in response to oxidative stress by governing the expression of genes encoding antioxidant enzymes that shield cells from diverse oxidative alterations. NRF2 and its negative regulator Kelch-like ECH-associated protein 1 (KEAP1) have been the focus of numerous investigations in elucidating whether NRF2 suppresses tumor promotion or conversely exerts pro-oncogenic effects. NRF2 has been found to participate in various pathological processes, including dysregulated cell proliferation, metabolic remodeling, and resistance to apoptosis. Herein, this review article will examine the intriguing role of phase separation in activating the NRF2 transcriptional activity and explore the NRF2 dual impacts on tumor immunology, cancer stem cells, metastasis, and long non-coding RNAs (LncRNAs). Taken together, this review aims to discuss the NRF2 multifaceted roles in both cancer prevention and promotion while also addressing the advantages, disadvantages, and limitations associated with modulating NRF2 therapeutically in cancer treatment.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Cinthya Enríquez
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
- Programa de Magister en Ciencias Químicas y Farmacéuticas, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile
| | - Constanza González
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
| | - Gabriela Aguirre-Martínez
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
- Instituto de Química Medicinal, Universidad Arturo Prat, Iquique 1100000, Chile
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
- Instituto de Química Medicinal, Universidad Arturo Prat, Iquique 1100000, Chile
- Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|
144
|
Yeung SHS, Lee RHS, Cheng GWY, Ma IWT, Kofler J, Kent C, Ma F, Herrup K, Fornage M, Arai K, Tse KH. White matter hyperintensity genetic risk factor TRIM47 regulates autophagy in brain endothelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.18.566359. [PMID: 38187529 PMCID: PMC10769267 DOI: 10.1101/2023.12.18.566359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
White matter hyperintensity (WMH) is strongly correlated with age-related dementia and hypertension, but its pathogenesis remains obscure. GWAS identified TRIM47 at 17q25 locus as a top genetic risk factor for WMH formation. TRIM family is a class of E3 ubiquitin ligase with pivotal functions in autophagy, which is critical for brain endothelial cell (ECs) remodeling during hypertension. We hypothesize that TRIM47 regulates autophagy and its loss-of-function disturbs cerebrovasculature. Based on transcriptomics and immunohistochemistry, TRIM47 is found selectively expressed by brain ECs in human and mouse, and its transcription is upregulated by artificially-induced autophagy while downregulated in hypertension-like conditions. Using in silico simulation, immunocytochemistry and super-resolution microscopy, we identified the highly conserved binding site between TRIM47 and the LIR (LC3-interacting region) motif of LC3B. Importantly, pharmacological autophagy induction increased Trim47 expression on mouse ECs (b.End3) culture, while silencing Trim47 significantly increased autophagy with ULK1 phosphorylation induction, transcription and vacuole formation. Together, we confirm that TRIM47 is an endogenous inhibitor of autophagy in brain ECs, and such TRIM47-mediated regulation connects genetic and physiological risk factors for WMH formation but warrants further investigation. SUMMARY STATEMENT TRIM47, top genetic risk factor for white matter hyperintensity formation, is a negative regulator of autophagy in brain endothelial cells and implicates a novel cellular mechanism for age-related cerebrovascular changes.
Collapse
|
145
|
Cirotti C, Taddei I, Contadini C, Di Girolamo C, Pepe G, De Bardi M, Borsellino G, Helmer-Citterich M, Barilà D. NRF2 connects Src tyrosine kinase to ferroptosis resistance in glioblastoma. Life Sci Alliance 2024; 7:e202302205. [PMID: 37879937 PMCID: PMC10599979 DOI: 10.26508/lsa.202302205] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023] Open
Abstract
Glioblastoma is a severe brain tumor characterized by an extremely poor survival rate of patients. Glioblastoma cancer cells escape to standard therapeutic protocols consisting of a combination of ionizing radiation and temozolomide alkylating drugs that trigger DNA damage by rewiring of signaling pathways. In recent years, the up-regulation of factors that counteract ferroptosis has been highlighted as a major driver of cancer resistance to ionizing radiation, although the molecular connection between the activation of oncogenic signaling and the modulation of ferroptosis has not been clarified yet. Here, we provide the first evidence for a molecular connection between the constitutive activation of tyrosine kinases and resistance to ferroptosis. Src tyrosine kinase, a central hub on which deregulated receptor tyrosine kinase signaling converge in cancer, leads to the stabilization and activation of NRF2 pathway, thus promoting resistance to ionizing radiation-induced ferroptosis. These data suggest that the up-regulation of the Src-NRF2 axis may represent a vulnerability for combined strategies that, by targeting ferroptosis resistance, enhance radiation sensitivity in glioblastoma.
Collapse
Affiliation(s)
- Claudia Cirotti
- Department of Biology, University of Rome "Tor Vergata," Rome, Italy
- Laboratory of Cell Signaling, IRCCS-Fondazione Santa Lucia, Rome, Italy
| | - Irene Taddei
- Department of Biology, University of Rome "Tor Vergata," Rome, Italy
- Laboratory of Cell Signaling, IRCCS-Fondazione Santa Lucia, Rome, Italy
| | - Claudia Contadini
- Department of Biology, University of Rome "Tor Vergata," Rome, Italy
- Laboratory of Cell Signaling, IRCCS-Fondazione Santa Lucia, Rome, Italy
- UOSD Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Gerardo Pepe
- Department of Biology, University of Rome "Tor Vergata," Rome, Italy
| | - Marco De Bardi
- Neuroimmunology Unit, Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giovanna Borsellino
- Neuroimmunology Unit, Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | - Daniela Barilà
- Department of Biology, University of Rome "Tor Vergata," Rome, Italy
- Laboratory of Cell Signaling, IRCCS-Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
146
|
Chao X, Niu M, Wang S, Ma X, Yang X, Sun H, Hu X, Wang H, Zhang L, Huang R, Xia M, Ballabio A, Jaeschke H, Ni HM, Ding WX. High-throughput screening of novel TFEB agonists in protecting against acetaminophen-induced liver injury in mice. Acta Pharm Sin B 2024; 14:190-206. [PMID: 38261809 PMCID: PMC10793101 DOI: 10.1016/j.apsb.2023.10.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 01/25/2024] Open
Abstract
Macroautophagy (referred to as autophagy hereafter) is a major intracellular lysosomal degradation pathway that is responsible for the degradation of misfolded/damaged proteins and organelles. Previous studies showed that autophagy protects against acetaminophen (APAP)-induced injury (AILI) via selective removal of damaged mitochondria and APAP protein adducts. The lysosome is a critical organelle sitting at the end stage of autophagy for autophagic degradation via fusion with autophagosomes. In the present study, we showed that transcription factor EB (TFEB), a master transcription factor for lysosomal biogenesis, was impaired by APAP resulting in decreased lysosomal biogenesis in mouse livers. Genetic loss-of and gain-of function of hepatic TFEB exacerbated or protected against AILI, respectively. Mechanistically, overexpression of TFEB increased clearance of APAP protein adducts and mitochondria biogenesis as well as SQSTM1/p62-dependent non-canonical nuclear factor erythroid 2-related factor 2 (NRF2) activation to protect against AILI. We also performed an unbiased cell-based imaging high-throughput chemical screening on TFEB and identified a group of TFEB agonists. Among these agonists, salinomycin, an anticoccidial and antibacterial agent, activated TFEB and protected against AILI in mice. In conclusion, genetic and pharmacological activating TFEB may be a promising approach for protecting against AILI.
Collapse
Affiliation(s)
- Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Mengwei Niu
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Shaogui Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiaowen Ma
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiao Yang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hua Sun
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Xujia Hu
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hua Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Li Zhang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples 80131, Italy
- Medical Genetics, Department of Translational Medicine, Federico II University, Naples 80131, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Internal Medicine, Division of Gastroenterology, Hepatology & Motility, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
147
|
Xing Z, Jiang X, Wu Y, Yu Z. Targeted Mevalonate Pathway and Autophagy in Antitumor Immunotherapy. Curr Cancer Drug Targets 2024; 24:890-909. [PMID: 38275055 DOI: 10.2174/0115680096273730231206054104] [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: 08/17/2023] [Revised: 09/30/2023] [Accepted: 10/11/2023] [Indexed: 01/27/2024]
Abstract
Tumors of the digestive system are currently one of the leading causes of cancer-related death worldwide. Despite considerable progress in tumor immunotherapy, the prognosis for most patients remains poor. In the tumor microenvironment (TME), tumor cells attain immune escape through immune editing and acquire immune tolerance. The mevalonate pathway and autophagy play important roles in cancer biology, antitumor immunity, and regulation of the TME. In addition, there is metabolic crosstalk between the two pathways. However, their role in promoting immune tolerance in digestive system tumors has not previously been summarized. Therefore, this review focuses on the cancer biology of the mevalonate pathway and autophagy, the regulation of the TME, metabolic crosstalk between the pathways, and the evaluation of their efficacy as targeted inhibitors in clinical tumor immunotherapy.
Collapse
Affiliation(s)
- Zongrui Xing
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Xiangyan Jiang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yuxia Wu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Zeyuan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| |
Collapse
|
148
|
Arabnezhad MR, Haghani F, Ghaffarian-Bahraman A, Jafarzadeh E, Mohammadi H, Yadegari JG, Farkhondeh T, Aschner M, Darroudi M, Marouzi S, Samarghandian S. Involvement of Nrf2 Signaling in Lead-induced Toxicity. Curr Med Chem 2024; 31:3529-3549. [PMID: 37221680 DOI: 10.2174/0929867330666230522143341] [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: 12/10/2022] [Revised: 03/19/2023] [Accepted: 03/31/2023] [Indexed: 05/25/2023]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is used as one of the main protective factors against various pathological processes, as it regulates cells resistant to oxidation. Several studies have extensively explored the relationship between environmental exposure to heavy metals, particularly lead (Pb), and the development of various human diseases. These metals have been reported to be able to, directly and indirectly, induce the production of reactive oxygen species (ROS) and cause oxidative stress in various organs. Since Nrf2 signaling is important in maintaining redox status, it has a dual role depending on the specific biological context. On the one hand, Nrf2 provides a protective mechanism against metal-induced toxicity; on the other hand, it can induce metalinduced carcinogenesis upon prolonged exposure and activation. Therefore, the aim of this review was to summarize the latest knowledge on the functional interrelation between toxic metals, such as Pb and Nrf2 signaling.
Collapse
Affiliation(s)
- Mohammad-Reza Arabnezhad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Haghani
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Ghaffarian-Bahraman
- Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Emad Jafarzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hamidreza Mohammadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Javad Ghasemian Yadegari
- Department of Pharmacognosy, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Majid Darroudi
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Somayeh Marouzi
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran
| |
Collapse
|
149
|
Ryan L, Rubinsztein DC. The autophagy of stress granules. FEBS Lett 2024; 598:59-72. [PMID: 38101818 DOI: 10.1002/1873-3468.14787] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/20/2023] [Accepted: 11/10/2023] [Indexed: 12/17/2023]
Abstract
Our understanding of stress granule (SG) biology has deepened considerably in recent years, and with this, increased understanding of links has been made between SGs and numerous neurodegenerative diseases. One of the proposed mechanisms by which SGs and any associated protein aggregates may become pathological is based upon defects in their autophagic clearance, and so the precise processes governing the degradation of SGs are important to understand. Mutations and disease-associated variants implicated in amyotrophic lateral sclerosis, Huntington's disease, Parkinson's disease and frontotemporal lobar dementia compromise autophagy, whilst autophagy-inhibiting drugs or knockdown of essential autophagy proteins result in the persistence of SGs. In this review, we will consider the current knowledge regarding the autophagy of SG.
Collapse
Affiliation(s)
- Laura Ryan
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR), University of Cambridge, UK
| |
Collapse
|
150
|
Danieli A, Vucak G, Baccarini M, Martens S. Sequestration of translation initiation factors in p62 condensates. Cell Rep 2023; 42:113583. [PMID: 38096057 DOI: 10.1016/j.celrep.2023.113583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 10/20/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
Selective autophagy mediates the removal of harmful material from the cytoplasm. This cargo material is selected by cargo receptors, which orchestrate its sequestration within double-membrane autophagosomes and subsequent lysosomal degradation. The cargo receptor p62/SQSTM1 is present in cytoplasmic condensates, and a fraction of them are constantly delivered into lysosomes. However, the molecular composition of the p62 condensates is incompletely understood. To obtain insights into their composition, we develop a method to isolate these condensates and find that p62 condensates are enriched in components of the translation machinery. Furthermore, p62 interacts with translation initiation factors, and eukaryotic initiation factor 2α (eIF2α) and eIF4E are degraded by autophagy in a p62-dependent manner. Thus, p62-mediated autophagy may in part be linked to down-regulation of translation initiation. The p62 condensate isolation protocol developed here may facilitate the study of their contribution to cellular quality control and their roles in health and disease.
Collapse
Affiliation(s)
- Alberto Danieli
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria.
| | - Georg Vucak
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Manuela Baccarini
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Sascha Martens
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria.
| |
Collapse
|