1
|
Saeed BI, Uthirapathy S, Kubaev A, Ganesan S, Shankhyan A, Gupta S, Joshi KK, Kariem M, Jasim AS, Ahmed JK. Ferroptosis as a key player in the pathogenesis and intervention therapy in liver injury: focusing on drug-induced hepatotoxicity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04115-w. [PMID: 40244448 DOI: 10.1007/s00210-025-04115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Globally, drug-induced hepatotoxicity or drug-induced liver injury (DILI) is a serious clinical concern. Knowing the processes and patterns of cell death is essential for finding new therapeutic targets since there are not many alternatives to therapy for severe liver lesions. Excessive lipid peroxidation is a hallmark of ferroptosis, an iron-reliant non-apoptotic cell death linked to various liver pathologies. When iron is pathogenic, concomitant inflammation may exacerbate iron-mediated liver injury, and the hepatocyte necrosis that results is a key element in the fibrogenic response. The idea that dysregulated metabolic pathways and compromised iron homeostasis contribute to the development of liver injury by ferroptosis is being supported by new data. Various ferroptosis-linked genes and pathways have been linked to liver injury, although the molecular processes behind ferroptosis's pathogenicity are not well known. Here, we delve into the features of ferroptosis, the processes governing ferroptosis, and our current knowledge of iron metabolism. We also provide an overview of ferroptosis's involvement in the pathophysiology of liver injury, particularly DILI. Lastly, the therapeutic possibilities of ferroptosis targeting for liver injury management have been provided. Natural products, nanoparticles (NPs), mesenchymal stem cell (MSC), and their exosomes have attracted increasing attention among such therapeutics.
Collapse
Affiliation(s)
- Bahaa Ibrahim Saeed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India
- Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
| | - Ahmed Salman Jasim
- Radiology Techniques Department College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Jawad Kadhim Ahmed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
2
|
Zhao Y, Gao L, Chen J, Wei J, Lin G, Hu K, Zhao W, Wei W, Huang W, Gao L, Yuan A, Qian K, Chen AF, Pu J. Remote limb ischemic conditioning alleviates steatohepatitis via extracellular vesicle-mediated muscle-liver crosstalk. Cell Metab 2025; 37:886-902.e7. [PMID: 40118054 DOI: 10.1016/j.cmet.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 08/30/2024] [Accepted: 02/25/2025] [Indexed: 03/23/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is an advanced form of liver disease with adverse outcomes. Manipulating interorgan communication is considered a promising strategy for managing metabolic disease, including steatohepatitis. Here, we report that remote limb ischemic conditioning (RIC), a clinically validated therapy for distant organ protection by transient muscle ischemia, significantly alleviated steatohepatitis in different mouse models. The beneficial effect of limb ischemic conditioning was mediated by muscle-to-liver transfer of small extracellular vesicles (sEVs) and their cargo microRNAs, leading to elevation of miR-181d-5p in the liver. Hepatic miR-181d-5p overexpression faithfully mirrored the molecular and histological benefits of limb ischemic conditioning by suppressing nuclear receptor 4A3 (NR4A3). Furthermore, circulating EVs from human volunteers undergoing limb ischemic conditioning improved steatohepatitis and transcriptomic perturbations in primary human hepatocytes and animal models. Our data underscore the translational potential of limb ischemic conditioning for steatohepatitis management and extend our understanding of muscle-liver crosstalk.
Collapse
Affiliation(s)
- Yichao Zhao
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Gao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Jianqing Chen
- Graduate School of Bengbu Medical College, Bengbu, Anhui, China
| | - Jingze Wei
- Graduate School of Bengbu Medical College, Bengbu, Anhui, China
| | - Guanqiao Lin
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kewei Hu
- Graduate School of Bengbu Medical College, Bengbu, Anhui, China
| | - Wubin Zhao
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Huang
- Department of Radiation Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingchen Gao
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ancai Yuan
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Qian
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; School of Biomedical Engineering, Institute of Medical Robotics and Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Graduate School of Bengbu Medical College, Bengbu, Anhui, China.
| |
Collapse
|
3
|
Etemad A, Tanaka Y, Wang S, Slae M, Sultan M, Elpeleg O, Hirokawa N. Mutations in the kinesin KIF12 promote MASH in humans and mice by disrupting lipogenic enzyme turnover. EMBO J 2025; 44:1608-1640. [PMID: 39920308 PMCID: PMC11914266 DOI: 10.1038/s44318-025-00366-8] [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: 05/14/2024] [Revised: 12/22/2024] [Accepted: 01/08/2025] [Indexed: 02/09/2025] Open
Abstract
As a common cause of liver cirrhosis, metabolic dysfunction-associated steatohepatitis (MASH) is regarded as a target of therapeutic intervention. However, a successful therapy has not yet been found, partly because the molecular pathogenesis is largely elusive. Here we show that KIF12 kinesin suppresses MASH development by accelerating the breakdown of two lipid biosynthesis enzymes, acetyl-CoA carboxylase 1 (ACC1) and pyruvate carboxylase (PC), in hepatocytes. We report three familial early-onset liver cirrhosis pedigrees with homozygous KIF12 mutations, accompanying MASH-like steatosis and cholestasis. The mouse genetic model carrying the corresponding Kif12 nonsense mutation faithfully reproduced the phenotypes as early as between 8 and 10 weeks of age. Furthermore, KIF12-deficient HepG2 cells exhibited significant steatosis, which was ameliorated by overexpressing a proline-rich domain (PRD) of KIF12. We found that KIF12-PRD promotes the degradation of ACC1 and PC, and this effect is likely to be through its direct interaction with these enzymes. Interestingly, KIF12 enhanced the ubiquitination of ACC1 by the E3 ligase COP1 and colocalized with these proteins as seen by super-resolution microscopy imaging. These data propose a role for KIF12 in suppressing MASH by accelerating turnover of lipogenic enzymes.
Collapse
Affiliation(s)
- Asieh Etemad
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo, 113-0033, Japan
| | - Yosuke Tanaka
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo, 113-0033, Japan
| | - Shuo Wang
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo, 113-0033, Japan
| | - Mordechai Slae
- Department of Genetics, Hadassah Hebrew University Medical Center, Jerusalem, 91120, Israel
| | - Mutaz Sultan
- Makassed Hospital, Faculty of Medicine, Al-Quds University, Jerusalem, Palestine
| | - Orly Elpeleg
- Department of Genetics, Hadassah Hebrew University Medical Center, Jerusalem, 91120, Israel
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo, 113-0033, Japan.
- Department of Advanced Morphological Imaging, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
4
|
Devasia AG, Ramasamy A, Leo CH. Current Therapeutic Landscape for Metabolic Dysfunction-Associated Steatohepatitis. Int J Mol Sci 2025; 26:1778. [PMID: 40004240 PMCID: PMC11855529 DOI: 10.3390/ijms26041778] [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: 11/07/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
In recent years, "metabolic dysfunction-associated steatotic liver disease" (MASLD) has been proposed to better connect liver disease to metabolic dysfunction, which is the most common chronic liver disease worldwide. MASLD affects more than 30% of individuals globally, and it is diagnosed by the combination of hepatic steatosis and obesity, type 2 diabetes, or two metabolic risk factors. MASLD begins with the buildup of extra fat, often greater than 5%, within the liver, causing liver hepatocytes to become stressed. This can proceed to a more severe form, metabolic dysfunction-associated steatohepatitis (MASH), in 20-30% of people, where inflammation in the liver causes tissue fibrosis, which limits blood flow over time. As fibrosis worsens, MASH may lead to cirrhosis, liver failure, or even liver cancer. While the pathophysiology of MASLD is not fully known, the current "multiple-hits" concept proposes that dietary and lifestyle factors, metabolic factors, and genetic or epigenetic factors contribute to elevated oxidative stress and inflammation, causing liver fibrosis. This review article provides an overview of the pathogenesis of MASLD and evaluates existing therapies as well as pharmacological drugs that are currently being studied in clinical trials for MASLD or MASH.
Collapse
Affiliation(s)
- Arun George Devasia
- Science, Math & Technology, Singapore University of Technology & Design, Singapore 487372, Singapore;
- Genome Institute of Singapore (GIS), Agency for Science Technology and Research (A*STAR), 60 Biopolis Street, Singapore 138672, Singapore;
| | - Adaikalavan Ramasamy
- Genome Institute of Singapore (GIS), Agency for Science Technology and Research (A*STAR), 60 Biopolis Street, Singapore 138672, Singapore;
| | - Chen Huei Leo
- Department of Biomedical Engineering, College of Design & Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| |
Collapse
|
5
|
Upadhyay G, Gowda SGB, Mishra SP, Nath LR, James A, Kulkarni A, Srikant Y, Upendram R, Marimuthu M, Hui SP, Jain S, Vasundhara K, Yadav H, Halade GV. Targeted and untargeted lipidomics with integration of liver dynamics and microbiome after dietary reversal of obesogenic diet targeting inflammation-resolution signaling in aging mice. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159542. [PMID: 39097080 DOI: 10.1016/j.bbalip.2024.159542] [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: 12/20/2023] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Obesity, a global epidemic linked to around 4 million deaths yearly, arises from lifestyle imbalances impacting inflammation-related conditions like non-alcoholic fatty liver disease and gut dysbiosis. But the long-term effects of inflammation caused by lifestyle-related dietary changes remain unexplained. In this study, we used young male C57Bl/6 mice which were fed either an obesogenic diet (OBD) or a control diet (CON) for six months. Later, a group of mice from the OBD group were intervened to the CON diet (OBD-R) for four months, while another OBD group remained on the OBD diet. The OBD induced distinct changes in gut microbial, notably elevating Firmicutes and Actinobacteria, while reducing Bacteroidetes and Tenericutes. OBD-R restored microbial abundance like CON. Analyzing liver, plasma, and fecal samples revealed OBD-induced alterations in various structural and bioactive lipids, which were normalized to CON in the OBD-R, showcasing lipid metabolism flexibility and adaptability to dietary shifts. OBD increased omega 6 fatty acid, Arachidonic Acid (AA) and decreased omega 3-derived lipid mediators in the OBD mimicking non-alcoholic fatty liver disease thus impacting inflammation-resolution pathways. OBD also induced hepatic inflammation via increasing alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels and proinflammatory markers CCR2, TNF-α, and IL-1β in liver. Transitioning from OBD to CON mitigated inflammatory gene expression and restored lipid and cholesterol networks. This study underscores the intricate interplay between lifestyle-driven dietary changes, gut microbiota, lipid metabolism, and liver health. Notably, it suggests that shift from an OBD (omega-6 enriched) to CON partially alleviates signs of chronic inflammation during aging. Understanding these microbial, lipidomic, and hepatic inflammatory dynamics reveals potential therapeutic avenues for metabolic disorders induced by diet, emphasizing the pivotal role of diet in sustaining metabolic health.
Collapse
Affiliation(s)
- Gunjan Upadhyay
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Siddabasave Gowda B Gowda
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan; Graduate School of Global Food Resources, Hokkaido University, Sapporo, Japan
| | - Sidharth P Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA
| | - Lipsa Rani Nath
- Graduate School of Global Food Resources, Hokkaido University, Sapporo, Japan
| | - Adewale James
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA
| | - Alisha Kulkarni
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Yuktee Srikant
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Rohitram Upendram
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - MathanKumar Marimuthu
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA
| | - Kain Vasundhara
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA; Center for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Ganesh V Halade
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
6
|
Dewald Z, Adesanya O, Bae H, Gupta A, Derham JM, Chembazhi UV, Kalsotra A. Altered drug metabolism and increased susceptibility to fatty liver disease in a mouse model of myotonic dystrophy. Nat Commun 2024; 15:9062. [PMID: 39433769 PMCID: PMC11494077 DOI: 10.1038/s41467-024-53378-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024] Open
Abstract
Myotonic Dystrophy type 1 (DM1), a highly prevalent form of muscular dystrophy, is caused by (CTG)n repeat expansion in the DMPK gene. Much of DM1 research has focused on the effects within the muscle and neurological tissues; however, DM1 patients also suffer from various metabolic and liver dysfunctions such as increased susceptibility to metabolic dysfunction-associated fatty liver disease (MAFLD) and heightened sensitivity to certain drugs. Here, we generated a liver-specific DM1 mouse model that reproduces molecular and pathological features of the disease, including susceptibility to MAFLD and reduced capacity to metabolize specific analgesics and muscle relaxants. Expression of CUG-expanded (CUG)exp repeat RNA within hepatocytes sequestered muscleblind-like proteins and triggered widespread gene expression and RNA processing defects. Mechanistically, we demonstrate that increased expression and alternative splicing of acetyl-CoA carboxylase 1 drives excessive lipid accumulation in DM1 livers, which is exacerbated by high-fat, high-sugar diets. Together, these findings reveal that (CUG)exp RNA toxicity disrupts normal hepatic functions, predisposing DM1 livers to injury, MAFLD, and drug clearance pathologies that may jeopardize the health of affected individuals and complicate their treatment.
Collapse
Affiliation(s)
- Zachary Dewald
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | | | - Haneui Bae
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Andrew Gupta
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Jessica M Derham
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Ullas V Chembazhi
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Auinash Kalsotra
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA.
- Cancer Center@Illinois, University of Illinois, Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois, Urbana-Champaign, Urbana, IL, USA.
- Chan Zuckerburg Biohub, Chicago, IL, USA.
| |
Collapse
|
7
|
Xie Z, Li Y, Cheng L, Huang Y, Rao W, Shi H, Li J. Potential therapeutic strategies for MASH: from preclinical to clinical development. LIFE METABOLISM 2024; 3:loae029. [PMID: 39872142 PMCID: PMC11749562 DOI: 10.1093/lifemeta/loae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/16/2024] [Accepted: 07/05/2024] [Indexed: 01/03/2025]
Abstract
Current treatment paradigms for metabolic dysfunction-associated steatohepatitis (MASH) are based primarily on dietary restrictions and the use of existing drugs, including anti-diabetic and anti-obesity medications. Given the limited number of approved drugs specifically for MASH, recent efforts have focused on promising strategies that specifically target hepatic lipid metabolism, inflammation, fibrosis, or a combination of these processes. In this review, we examined the pathophysiology underlying the development of MASH in relation to recent advances in effective MASH therapy. Particularly, we analyzed the effects of lipogenesis inhibitors, nuclear receptor agonists, glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists, fibroblast growth factor mimetics, and combinatorial therapeutic approaches. We summarize these targets along with their preclinical and clinical candidates with the ultimate goal of optimizing the therapeutic prospects for MASH.
Collapse
Affiliation(s)
- Zhifu Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yufeng Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Long Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yidan Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanglin Rao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Honglu Shi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingya Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| |
Collapse
|
8
|
Yang Y, Wu J, Zhou W, Ji G, Dang Y. Protein posttranslational modifications in metabolic diseases: basic concepts and targeted therapies. MedComm (Beijing) 2024; 5:e752. [PMID: 39355507 PMCID: PMC11442990 DOI: 10.1002/mco2.752] [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: 05/25/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024] Open
Abstract
Metabolism-related diseases, including diabetes mellitus, obesity, hyperlipidemia, and nonalcoholic fatty liver disease, are becoming increasingly prevalent, thereby posing significant threats to human health and longevity. Proteins, as the primary mediators of biological activities, undergo various posttranslational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, methylation, and SUMOylation, among others, which substantially diversify their functions. These modifications are crucial in the physiological and pathological processes associated with metabolic disorders. Despite advancements in the field, there remains a deficiency in contemporary summaries addressing how these modifications influence processes of metabolic disease. This review aims to systematically elucidate the mechanisms through which PTM of proteins impact the progression of metabolic diseases, including diabetes, obesity, hyperlipidemia, and nonalcoholic fatty liver disease. Additionally, the limitations of the current body of research are critically assessed. Leveraging PTMs of proteins provides novel insights and therapeutic targets for the prevention and treatment of metabolic disorders. Numerous drugs designed to target these modifications are currently in preclinical or clinical trials. This review also provides a comprehensive summary. By elucidating the intricate interplay between PTMs and metabolic pathways, this study advances understanding of the molecular mechanisms underlying metabolic dysfunction, thereby facilitating the development of more precise and effective disease management strategies.
Collapse
Affiliation(s)
- Yunuo Yang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Jiaxuan Wu
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Wenjun Zhou
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Guang Ji
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Yanqi Dang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| |
Collapse
|
9
|
Bali AD, Rosenzveig A, Frishman WH, Aronow WS. Nonalcoholic Fatty Liver Disease and Cardiovascular Disease: Causation or Association. Cardiol Rev 2024; 32:453-462. [PMID: 36825899 DOI: 10.1097/crd.0000000000000537] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a disease process that is gaining increasing recognition. The global prevalence of NAFLD is increasing in parallel with growing rates of risk factors for NAFLD such as hypertension, obesity, diabetes, and metabolic syndrome. NAFLD has been referred to as a risk factor for cardiovascular disease (CVD). As CVD is the leading cause of morbidity and mortality worldwide, there are constant efforts to describe and alleviate its risk factors. Although there is conflicting data supporting NAFLD as a causative or associative factor for CVD, NAFLD has been shown to be associated with structural, electrical, and atherosclerotic disease processes of the heart. Shared risk factors and pathophysiologic mechanisms between NAFLD and CVD warrant further explication. Pathologic mechanisms such as endothelial dysfunction, oxidative stress, insulin resistance, genetic underpinnings, and gut microbiota dysregulation have been described in both CVD and NAFLD. The mainstay of treatment for NAFLD is lifestyle intervention including physical exercise and hypocaloric intake in addition to bariatric surgery. Investigations into various therapeutic targets to alleviate hepatic steatosis and fibrosis by way of maintaining the balance between lipid synthesis and breakdown. A major obstacle preventing the success of many pharmacologic approaches has been the effects of these medications on CVD risk. The future of pharmacologic treatment of NAFLD is promising as effective medications with limited CVD harm are being investigated.
Collapse
Affiliation(s)
- Atul D Bali
- From the Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | | | - William H Frishman
- From the Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Wilbert S Aronow
- From the Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
10
|
Escuder-Rodríguez JJ, Liang D, Jiang X, Sinicrope FA. Ferroptosis: Biology and Role in Gastrointestinal Disease. Gastroenterology 2024; 167:231-249. [PMID: 38431204 PMCID: PMC11193643 DOI: 10.1053/j.gastro.2024.01.051] [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: 07/28/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/05/2024]
Abstract
Ferroptosis is a form of nonapoptotic cell death that involves iron-dependent phospholipid peroxidation induced by accumulation of reactive oxygen species, and results in plasma membrane damage and the release of damage-associated molecular patterns. Ferroptosis has been implicated in aging and immunity, as well as disease states including intestinal and liver conditions and cancer. To date, several ferroptosis-associated genes and pathways have been implicated in liver disease. Although ferroptotic cell death is associated with dysfunction of the intestinal epithelium, the underlying molecular basis is poorly understood. As the mechanisms regulating ferroptosis become further elucidated, there is clear potential to use ferroptosis to achieve therapeutic benefit.
Collapse
Affiliation(s)
- Juan-José Escuder-Rodríguez
- Department of Medicine, Gastrointestinal Research Unit, Mayo Clinic Alix School of Medicine, Rochester, Minnesota
| | - Deguang Liang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York.
| | - Frank A Sinicrope
- Department of Medicine, Gastrointestinal Research Unit, Mayo Clinic Alix School of Medicine, Rochester, Minnesota.
| |
Collapse
|
11
|
Yu Y, Liang J, Yuan Z, Wang A, Liu X, Chen Y, Zhang M, Gao Y, Zhang H, Liu Y. Bioactive compound schaftoside from Clinacanthus nutans attenuates acute liver injury by inhibiting ferroptosis through activation the Nrf2/GPX4 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118135. [PMID: 38556139 DOI: 10.1016/j.jep.2024.118135] [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: 01/22/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Clinacanthus nutans (Burm. f.) Lindau, a traditional herb renowned for its anti-tumor, antioxidant, and anti-inflammatory properties, has garnered considerable attention. Although its hepatoprotective effects have been described, there is still limited knowledge of its treatment of acute liver injury (ALI), and its mechanisms remain unclear. AIM OF THE STUDY To assess the efficacy of Clinacanthus nutans in ALI and to identify the most effective fractions and their underlying mechanism of action. METHODS Bioinformatics was employed to explore the underlying anti-hepatic injury mechanisms and active compounds of Clinacanthus nutans. The binding ability of schaftoside, a potential active ingredient in Clinacanthus nutans, to the core target nuclear factor E2-related factor 2 (Nrf2) was further determined by molecular docking. The role of schaftoside in improving histological abnormalities in the liver was observed by H&E and Masson's staining in an ALI model induced by CCl4. Serum and liver biochemical parameters were measured using AST, ALT and hydroxyproline kits. An Fe2+ kit, transmission electron microscopy, western blotting, RT-qPCR, and DCFH-DA were used to measure whether schaftoside reduces ferroptosis-induced ALI. Subsequently, specific siRNA knockdown of Nrf2 in AML12 cells was performed to further elucidate the mechanism by which schaftoside attenuates ferroptosis-induced ALI. RESULTS Bioinformatics analysis and molecular docking showed that schaftoside is the principal compound from Clinacanthus nutans. Schaftoside was shown to diminish oxidative stress levels, attenuate liver fibrosis, and forestall ferroptosis. Deeper investigations revealed that schaftoside amplified Nrf2 expression and triggered the Nrf2/GPX4 pathway, thereby reversing mitochondrial aberrations triggered by lipid peroxidation, GPX4 depletion, and ferroptosis. CONCLUSION The lead compound schaftoside counters ferroptosis through the Nrf2/GPX4 axis, providing insights into a novel molecular mechanism for treating ALI, thereby presenting an innovative therapeutic strategy for ferroptosis-induced ALI.
Collapse
Affiliation(s)
- Yi Yu
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Jingwei Liang
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China; International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Haikou, 571199, China
| | - Zhexin Yuan
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Aiping Wang
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Xinxing Liu
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Yu Chen
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Min Zhang
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Yanan Gao
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China; International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Haikou, 571199, China
| | - Haiying Zhang
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China; International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Haikou, 571199, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, Haikou, 571199, China.
| | - Yan Liu
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China; International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Haikou, 571199, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, Haikou, 571199, China.
| |
Collapse
|
12
|
Pang Q, Tang Z, Luo L. The crosstalk between oncogenic signaling and ferroptosis in cancer. Crit Rev Oncol Hematol 2024; 197:104349. [PMID: 38626848 DOI: 10.1016/j.critrevonc.2024.104349] [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: 09/30/2023] [Revised: 03/13/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Ferroptosis, a novel form of cell death regulation, was identified in 2012. It is characterized by unique features that differentiate it from other types of cell death, including necrosis, apoptosis, autophagy, and pyroptosis. Ferroptosis is defined by an abundance of iron ions and lipid peroxidation, resulting in alterations in subcellular structures, an elevation in reactive oxygen species (ROS), a reduction in glutathione (GSH) levels, and an augmentation in Fe (II) cytokines. Ferroptosis, a regulated process, is controlled by an intricate network of signaling pathways, where multiple stimuli can either enhance or hinder the process. This review primarily examines the defensive mechanisms of ferroptosis and its interaction with the tumor microenvironment. The analysis focuses on the pathways that involve AMPK, p53, NF2, mTOR, System Xc-, Wnt, Hippo, Nrf2, and cGAS-STING. The text discusses the possibilities of employing a combination therapy that targets several pathways for the treatment of cancer. It emphasizes the necessity for additional study in this field.
Collapse
Affiliation(s)
- Qianghu Pang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Zhirou Tang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang,School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
13
|
Lin Y, Yang M, Huang L, Yang F, Fan J, Qiang Y, Chang Y, Zhou W, Yan L, Xiong J, Ping J, Chen S, Men D, Li F. A bacteria-derived tetramerized protein ameliorates nonalcoholic steatohepatitis in mice via binding and relocating acetyl-coA carboxylase. Cell Rep 2023; 42:113453. [PMID: 37976162 DOI: 10.1016/j.celrep.2023.113453] [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: 04/25/2023] [Revised: 09/30/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
Increased de novo lipogenesis (DNL) is a major feature of nonalcoholic steatohepatitis (NASH). None of the drugs targeting the catalytic activity of acetyl-CoA carboxylase (ACC), the rate-limiting enzyme in the DNL process, have been approved by the FDA. Whether cytosolic ACC1 can be regulated spatially remains to be explored. Herein, we find that streptavidin (SA), which is a bacterium-derived tetrameric protein, forms cytosolic condensates and efficiently induces a spatial re-localization of ACC1 in liver cells, concomitant with inhibited lipid accumulation. Both SA tetrameric structure and multivalent protein interaction are required for condensate formation. Interestingly, the condensates are further characterized as gel-like membraneless organelle (SAGMO) and significantly restrict the cytosolic dispersion of ACC1 and fatty acid synthase. Notably, AAV-mediated delivery of SA partially blocks mouse liver DNL and ameliorates NASH without eliciting hypertriglyceridemia. In summary, our study shows that insulating lipogenesis-related proteins by SAGMO might be effective for NASH treatment.
Collapse
Affiliation(s)
- Yan Lin
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Mingkun Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Li Huang
- Research Center for Medicine and Structural Biology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Fan Yang
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Jiachen Fan
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Yulong Qiang
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Yuting Chang
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Wenjie Zhou
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Leilei Yan
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Jie Xiong
- Department of Immunology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Jie Ping
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Shizhen Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Dong Men
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China.
| | - Feng Li
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Allergy and Immunology, Wuhan 430071, China.
| |
Collapse
|
14
|
Rao G, Peng X, Li X, An K, He H, Fu X, Li S, An Z. Unmasking the enigma of lipid metabolism in metabolic dysfunction-associated steatotic liver disease: from mechanism to the clinic. Front Med (Lausanne) 2023; 10:1294267. [PMID: 38089874 PMCID: PMC10711211 DOI: 10.3389/fmed.2023.1294267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/26/2023] [Indexed: 06/21/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly defined as non-alcoholic fatty liver disease (NAFLD), is a disorder marked by the excessive deposition of lipids in the liver, giving rise to a spectrum of liver pathologies encompassing steatohepatitis, fibrosis/cirrhosis, and hepatocellular carcinoma. Despite the alarming increase in its prevalence, the US Food and Drug Administration has yet to approve effective pharmacological therapeutics for clinical use. MASLD is characterized by the accretion of lipids within the hepatic system, arising from a disarray in lipid provision (whether through the absorption of circulating lipids or de novo lipogenesis) and lipid elimination (via free fatty acid oxidation or the secretion of triglyceride-rich lipoproteins). This disarray leads to the accumulation of lipotoxic substances, cellular pressure, damage, and fibrosis. Indeed, the regulation of the lipid metabolism pathway is intricate and multifaceted, involving a myriad of factors, such as membrane transport proteins, metabolic enzymes, and transcription factors. Here, we will review the existing literature on the key process of lipid metabolism in MASLD to understand the latest progress in this molecular mechanism. Notably, de novo lipogenesis and the roles of its two main transcription factors and other key metabolic enzymes are highlighted. Furthermore, we will delve into the realm of drug research, examining the recent progress made in understanding lipid metabolism in MASLD. Additionally, we will outline prospective avenues for future drug research on MASLD based on our unique perspectives.
Collapse
Affiliation(s)
- Guocheng Rao
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Xi Peng
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Department of Endocrinology and Metabolism, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, China
| | - Xinqiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Kang An
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, Multimorbidity Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - He He
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xianghui Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Shuangqing Li
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, Multimorbidity Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenmei An
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Guo S, Zhang BB, Gao L, Yu XY, Shen JH, Yang F, Zhang WC, Jin YG, Li G, Wang YG, Han ZY, Liu Y. RNF13 protects against pathological cardiac hypertrophy through p62-NRF2 pathway. Free Radic Biol Med 2023; 209:252-264. [PMID: 37852547 DOI: 10.1016/j.freeradbiomed.2023.10.395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 10/20/2023]
Abstract
Heart failure (HF) severely impairs human health because of its high incidence and mortality. Cardiac hypertrophy is the main cause of HF, while its underlying mechanism is not fully clear. As an E3 ubiquitin ligase, Ring finger protein 13 (RNF13) plays a crucial role in many disorders, such as liver immune, neurological disease and tumorigenesis, whereas the function of RNF13 in cardiac hypertrophy remains largely unknown. In the present study, we found that the protein expression of RNF13 is up-regulated in the transverse aortic constriction (TAC)-induced murine hypertrophic hearts and phenylephrine (PE)-induced cardiomyocyte hypertrophy. Functional investigations indicated that RNF13 global knockout mice accelerates the degree of TAC-induced cardiac hypertrophy, including cardiomyocyte enlargement, cardiac fibrosis and heart dysfunction. On the contrary, adeno-associated virus 9 (AAV9) mediated-RNF13 overexpression mice alleviated cardiac hypertrophy. Furthermore, we demonstrated that adenoviral RNF13 attenuates the PE-induced cardiomyocyte hypertrophy and down-regulates the expression of cardiac hypertrophic markers, while the opposite results were observed in the RNF13 knockdown group. The RNA-sequence of RNF13 knockout and wild type mice showed that RNF13 deficiency activates oxidative stress after TAC surgery. In terms of the mechanism, we found that RNF13 directly interacted with p62 and promoted the activation of downstream NRF2/HO-1 signaling. Finally, we proved that p62 knockdown can reverse the effect of RNF13 in cardiac hypertrophy. In conclusion, RNF13 protects against the cardiac hypertrophy via p62-NRF2 axis.
Collapse
Affiliation(s)
- Sen Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China.
| | - Bin-Bin Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Lu Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Xiao-Yue Yu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Ji-Hong Shen
- Department of Electrocardiogram, The Second Affiliated Hospital of Zhengzhou University, No.2 Jingba Road, Zhengzhou, China
| | - Fan Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Wen-Cai Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Ya-Ge Jin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Gang Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Yan-Ge Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Zhan-Ying Han
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China.
| | - Yuan Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China.
| |
Collapse
|
16
|
Sun S, Shen J, Jiang J, Wang F, Min J. Targeting ferroptosis opens new avenues for the development of novel therapeutics. Signal Transduct Target Ther 2023; 8:372. [PMID: 37735472 PMCID: PMC10514338 DOI: 10.1038/s41392-023-01606-1] [Citation(s) in RCA: 179] [Impact Index Per Article: 89.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/24/2023] [Accepted: 08/11/2023] [Indexed: 09/23/2023] Open
Abstract
Ferroptosis is an iron-dependent form of regulated cell death with distinct characteristics, including altered iron homeostasis, reduced defense against oxidative stress, and abnormal lipid peroxidation. Recent studies have provided compelling evidence supporting the notion that ferroptosis plays a key pathogenic role in many diseases such as various cancer types, neurodegenerative disease, diseases involving tissue and/or organ injury, and inflammatory and infectious diseases. Although the precise regulatory networks that underlie ferroptosis are largely unknown, particularly with respect to the initiation and progression of various diseases, ferroptosis is recognized as a bona fide target for the further development of treatment and prevention strategies. Over the past decade, considerable progress has been made in developing pharmacological agonists and antagonists for the treatment of these ferroptosis-related conditions. Here, we provide a detailed overview of our current knowledge regarding ferroptosis, its pathological roles, and its regulation during disease progression. Focusing on the use of chemical tools that target ferroptosis in preclinical studies, we also summarize recent advances in targeting ferroptosis across the growing spectrum of ferroptosis-associated pathogenic conditions. Finally, we discuss new challenges and opportunities for targeting ferroptosis as a potential strategy for treating ferroptosis-related diseases.
Collapse
Affiliation(s)
- Shumin Sun
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Shen
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianwei Jiang
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
17
|
Choi J, Choi H, Chung J. Icariin Supplementation Suppresses the Markers of Ferroptosis and Attenuates the Progression of Nonalcoholic Steatohepatitis in Mice Fed a Methionine Choline-Deficient Diet. Int J Mol Sci 2023; 24:12510. [PMID: 37569885 PMCID: PMC10419585 DOI: 10.3390/ijms241512510] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Icariin, a flavonoid abundant in the herb Epimedium, exhibits anti-ferroptotic activity. However, its impact on nonalcoholic steatohepatitis (NASH) development remains unclear. This study aimed to investigate the potential role of icariin in mitigating methionine choline-deficient (MCD) diet-induced NASH in C57BL/6J mice. The results showed that icariin treatment significantly reduced serum alanine aminotrasferase and aspartate aminotransferase activities while improving steatosis, inflammation, ballooning, and fibrosis in the liver tissues of mice fed the MCD diet. These improvements were accompanied by a substantial reduction in the hepatic iron contents and levels of malondialdehyde and 4-hydroxynonenal, as well as an increase in the activities of catalase and superoxide dismutase. Notably, icariin treatment suppressed the hepatic protein levels of ferroptosis markers such as acyl-CoA synthetase long-chain family member 4 and arachidonate 12-lipoxygenase, which were induced by the MCD diet. Furthermore, transmission electron microscopy confirmed the restoration of morphological changes in the mitochondria, a hallmark characteristic of ferroptosis, by icariin. Additionally, icariin treatment significantly increased the protein levels of Nrf2, a cystine/glutamate transporter (xCT), and glutathione peroxidase 4 (GPX4). In conclusion, our study suggests that icariin has the potential to attenuate NASH, possibly by suppressing ferroptosis via the Nrf2-xCT/GPX4 pathway.
Collapse
Affiliation(s)
| | | | - Jayong Chung
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea; (J.C.); (H.C.)
| |
Collapse
|
18
|
Qu P, Rom O, Li K, Jia L, Gao X, Liu Z, Ding S, Zhao M, Wang H, Chen S, Xiong X, Zhao Y, Xue C, Zhao Y, Chu C, Wen B, Finney AC, Zheng Z, Cao W, Zhao J, Bai L, Zhao S, Sun D, Zeng R, Lin J, Liu W, Zheng L, Zhang J, Liu E, Chen YE. DT-109 ameliorates nonalcoholic steatohepatitis in nonhuman primates. Cell Metab 2023; 35:742-757.e10. [PMID: 37040763 DOI: 10.1016/j.cmet.2023.03.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/03/2023] [Accepted: 03/17/2023] [Indexed: 04/13/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) prevalence is rising with no pharmacotherapy approved. A major hurdle in NASH drug development is the poor translatability of preclinical studies to safe/effective clinical outcomes, and recent failures highlight a need to identify new targetable pathways. Dysregulated glycine metabolism has emerged as a causative factor and therapeutic target in NASH. Here, we report that the tripeptide DT-109 (Gly-Gly-Leu) dose-dependently attenuates steatohepatitis and fibrosis in mice. To enhance the probability of successful translation, we developed a nonhuman primate model that histologically and transcriptionally mimics human NASH. Applying a multiomics approach combining transcriptomics, proteomics, metabolomics, and metagenomics, we found that DT-109 reverses hepatic steatosis and prevents fibrosis progression in nonhuman primates, not only by stimulating fatty acid degradation and glutathione formation, as found in mice, but also by modulating microbial bile acid metabolism. Our studies describe a highly translatable NASH model and highlight the need for clinical evaluation of DT-109.
Collapse
Affiliation(s)
- Pengxiang Qu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Oren Rom
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Ke Li
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Capital Medical University, 6 Tiantan Xili, Chongwen District, Beijing 100050, China
| | - Linying Jia
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Xiaojing Gao
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhipeng Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Shusi Ding
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Capital Medical University, 6 Tiantan Xili, Chongwen District, Beijing 100050, China
| | - Mingming Zhao
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 38 Xue Yuan Road, Beijing 100191, China
| | - Huiqing Wang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 38 Xue Yuan Road, Beijing 100191, China
| | - Shuangshuang Chen
- Department of Endocrinology and Metabolism, Fudan Institute of Metabolic Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Xuhui District, Shanghai 200031, China
| | - Xuelian Xiong
- Department of Endocrinology and Metabolism, Fudan Institute of Metabolic Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Xuhui District, Shanghai 200031, China
| | - Ying Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Chao Xue
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Yang Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Chengshuang Chu
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Zuowen Zheng
- Spring Biological Technology Development Co., Ltd, Fangchenggang, Guangxi 538000, China
| | - Wenbin Cao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Jinpeng Zhao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Liang Bai
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Sihai Zhao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rong Zeng
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiandie Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
| | - Lemin Zheng
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Capital Medical University, 6 Tiantan Xili, Chongwen District, Beijing 100050, China; The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, 38 Xue Yuan Road, Beijing 100191, China.
| | - Jifeng Zhang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI 48109, USA.
| | - Enqi Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi 710061, China.
| | - Y Eugene Chen
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI 48109, USA.
| |
Collapse
|
19
|
Wang X, Zhou Y, Min J, Wang F. Zooming in and out of ferroptosis in human disease. Front Med 2023; 17:173-206. [PMID: 37121959 DOI: 10.1007/s11684-023-0992-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/12/2023] [Indexed: 05/02/2023]
Abstract
Ferroptosis is defined as an iron-dependent regulated form of cell death driven by lipid peroxidation. In the past decade, it has been implicated in the pathogenesis of various diseases that together involve almost every organ of the body, including various cancers, neurodegenerative diseases, cardiovascular diseases, lung diseases, liver diseases, kidney diseases, endocrine metabolic diseases, iron-overload-related diseases, orthopedic diseases and autoimmune diseases. Understanding the underlying molecular mechanisms of ferroptosis and its regulatory pathways could provide additional strategies for the management of these disease conditions. Indeed, there are an expanding number of studies suggesting that ferroptosis serves as a bona-fide target for the prevention and treatment of these diseases in relevant pre-clinical models. In this review, we summarize the progress in the research into ferroptosis and its regulatory mechanisms in human disease, while providing evidence in support of ferroptosis as a target for the treatment of these diseases. We also discuss our perspectives on the future directions in the targeting of ferroptosis in human disease.
Collapse
Affiliation(s)
- Xue Wang
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ye Zhou
- Department of Endocrinology and Metabolism, Ningbo First Hospital, Ningbo, 315000, China
| | - Junxia Min
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Fudi Wang
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
20
|
Yang Z, Wang L. Current, emerging, and potential therapies for non-alcoholic steatohepatitis. Front Pharmacol 2023; 14:1152042. [PMID: 37063264 PMCID: PMC10097909 DOI: 10.3389/fphar.2023.1152042] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has been identified as the most common chronic liver disease worldwide, with a growing incidence. NAFLD is considered the hepatic manifestation of a metabolic syndrome that emerges from multiple factors (e.g., oxidative stress, metabolic disorders, endoplasmic reticulum stress, cell death, and inflammation). Non-alcoholic steatohepatitis (NASH), an advanced form of NAFLD, has been reported to be a leading cause of cirrhosis and hepatic carcinoma, and it is progressing rapidly. Since there is no approved pharmacotherapy for NASH, a considerable number of therapeutic targets have emerged with the deepening of the research on NASH pathogenesis. In this study, the therapeutic potential and properties of regulating metabolism, the gut microbiome, antioxidant, microRNA, inhibiting apoptosis, targeting ferroptosis, and stem cell-based therapy in NASH are reviewed and evaluated. Since the single-drug treatment of NASH is affected by individual heterogeneous responses and side effects, it is imperative to precisely carry out targeted therapy with low toxicity. Lastly, targeted therapeutic agent delivery based on exosomes is proposed in this study, such that drugs with different mechanisms can be incorporated to generate high-efficiency and low-toxicity individualized medicine.
Collapse
Affiliation(s)
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
21
|
Pirola CJ, Sookoian S. Advances in our understanding of the molecular heterogeneity of fatty liver disease: toward informed treatment decision making. Expert Rev Gastroenterol Hepatol 2023; 17:317-324. [PMID: 36912694 DOI: 10.1080/17474124.2023.2191190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
INTRODUCTION nonalcoholic fatty liver disease (NAFLD) is a complex disorder resulting from intricate relationships with diverse cardiometabolic risk factors and environmental factors. NAFLD may result in severe chronic liver damage and potentially declining liver function. AREAS COVERED Accumulated knowledge over the last decade indicates that the disease trajectory presents substantial heterogeneity. In addition, overlapping features with the diseases of the metabolic syndrome, combined with heterogeneity in disease mechanisms, further complicates NAFLD diagnosis and prognosis, and hampers progress in biomarker and pharmacological discoveries. Here, we explore solving the heterogeneous clinical landscape of NAFLD by cluster analysis of molecular signatures that serve as a proxy for disease stratification into molecular sub-types. First, we collected information on NAFLD and metabolic syndrome-associated protein-coding genes by data mining the literature. Next, we performed pathways enrichment and cluster analyses to decipher and dissect the different patterns of phenotypic heterogeneity. Our approach showed unique biological pathways for every clinical subtype/group, namely NAFLD + obesity, NAFLD + arterial hypertension, NAFLD + dyslipidemia, and NAFLD + type 2 diabetes. EXPERT OPINION Patients with NAFLD may be benefited by a better understanding of the disease biology, which involves 'dissection' of the molecular sub-phenotypes that drive the disease progression.
Collapse
Affiliation(s)
- Carlos J Pirola
- Systems Biology of Complex Diseases, Centro de Altos Estudios En Ciencias Humanas Y de la Salud (CAECIHS), Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Buenos Aires, Argentina
| | - Silvia Sookoian
- Clinical and Molecular Hepatology, Centro de Altos Estudios En Ciencias Humanas Y de la Salud (CAECIHS), Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
22
|
Tian R, Yang J, Wang X, Liu S, Dong R, Wang Z, Yang Z, Zhang Y, Cai Z, Yang H, Hu Y, She ZG, Li H, Zhou J, Zhang XJ. Honokiol acts as an AMPK complex agonist therapeutic in non-alcoholic fatty liver disease and metabolic syndrome. Chin Med 2023; 18:30. [PMID: 36932412 PMCID: PMC10024454 DOI: 10.1186/s13020-023-00729-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/15/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver (NAFLD) and its related metabolic syndrome have become major threats to human health, but there is still a need for effective and safe drugs to treat these conditions. Here we aimed to identify potential drug candidates for NAFLD and the underlying molecular mechanisms. METHODS A drug repositioning strategy was used to screen an FDA-approved drug library with approximately 3000 compounds in an in vitro hepatocyte model of lipid accumulation, with honokiol identified as an effective anti-NAFLD candidate. We systematically examined the therapeutic effect of honokiol in NAFLD and metabolic syndrome in multiple in vitro and in vivo models. Transcriptomic examination and biotin-streptavidin binding assays were used to explore the underlying molecular mechanisms, confirmed by rescue experiments. RESULTS Honokiol significantly inhibited metabolic syndrome and NAFLD progression as evidenced by improved hepatic steatosis, liver fibrosis, adipose inflammation, and insulin resistance. Mechanistically, the beneficial effects of honokiol were largely through AMPK activation. Rather than acting on the classical upstream regulators of AMPK, honokiol directly bound to the AMPKγ1 subunit to robustly activate AMPK signaling. Mutation of honokiol-binding sites of AMPKγ1 largely abolished the protective capacity of honokiol against NAFLD. CONCLUSION These findings clearly demonstrate the beneficial effects of honokiol in multiple models and reveal a previously unappreciated signaling mechanism of honokiol in NAFLD and metabolic syndrome. This study also provides new insights into metabolic disease treatment by targeting AMPKγ1 subunit-mediated signaling activation.
Collapse
Affiliation(s)
- Ruifeng Tian
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Jinjie Yang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Xiaoming Wang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Shuaiyang Liu
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Ruixiang Dong
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Zhenya Wang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Zifeng Yang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Yingping Zhang
- School of Pharmacy, Bengbu Medical College, Bengbu, 233030, China
| | - Zhiwei Cai
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Hailong Yang
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China
| | - Yufeng Hu
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China
| | - Zhi-Gang She
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.,Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Hongliang Li
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China. .,Institute of Model Animal of Wuhan University, Wuhan, 430071, China. .,Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China. .,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China. .,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Junjie Zhou
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China. .,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China.
| | - Xiao-Jing Zhang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China. .,Institute of Model Animal of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
23
|
Yu Y, Nie Q, Wang Z, Di Y, Chen X, Ren K. Targeting acetyl-CoA carboxylase 1 for cancer therapy. Front Pharmacol 2023; 14:1129010. [PMID: 36843935 PMCID: PMC9950103 DOI: 10.3389/fphar.2023.1129010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
Metabolic adaptation is an emerging hallmark of tumors. De novo fatty acid synthesis is an important metabolic process to produce metabolic intermediates for energy storage, biosynthesis of membrane lipids and generation of signaling molecules. Acetyl-CoA carboxylase 1 (ACC1) is a critical enzyme in the fatty acid synthesis, which carboxylates acetyl-CoA carboxylic acid to form malonyl-CoA. The role of acetyl-CoA carboxylase 1 in fatty acid synthesis makes it a promising therapeutic target for various metabolic diseases such as non-alcoholic fatty liver disease, obesity and diabetes. Tumors have a high energy flow and a strong dependence on fatty acid synthesis. Thus, acetyl-CoA carboxylase inhibition has become a potential choice for anti-tumor therapy. In this review, we first introduced the structure and expression pattern of Acetyl-CoA carboxylase 1. We also discussed the molecular mechanisms of acetyl-CoA carboxylase 1 in the initiation and progression of various cancer types. Furthermore, acetyl-CoA carboxylase1 inhibitors has also been discussed. Collectively, we summarized the interplay between acetyl-CoA carboxylase 1 and tumorigenesis, indicating acetyl-CoA carboxylase 1 as a promising therapeutic target for tumor management.
Collapse
Affiliation(s)
- Yong Yu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qingzhu Nie
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ziyi Wang
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yu Di
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaolong Chen
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Kaiming Ren
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
24
|
Lipidomics analysis in drug discovery and development. Curr Opin Chem Biol 2023; 72:102256. [PMID: 36586190 DOI: 10.1016/j.cbpa.2022.102256] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/08/2022] [Accepted: 11/28/2022] [Indexed: 12/30/2022]
Abstract
Despite being a relatively new addition to the Omics' landscape, lipidomics is increasingly being recognized as an important tool for the identification of druggable targets and biochemical markers. In this review we present recent advances of lipid analysis in drug discovery and development. We cover current state of the art technologies which are constantly evolving to meet demands in terms of sensitivity and selectivity. A careful selection of important examples is then provided, illustrating the versatility of lipidomics analysis in the drug discovery and development process. Integration of lipidomics with other omics', stem-cell technologies, and metabolic flux analysis will open new avenues for deciphering pathophysiological mechanisms and the discovery of novel targets and biomarkers.
Collapse
|
25
|
Wu A, Ye M, Ma T, She Z, Li R, Shi H, Yang L, Yi M, Li H. TBC1D25 alleviates nonalcoholic steatohepatitis by inhibiting abnormal lipid accumulation and inflammation. J Cell Physiol 2023; 238:393-406. [PMID: 36710714 DOI: 10.1002/jcp.30934] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 01/31/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a strong stimulant of cardiovascular diseases, affecting one-quarter of the world's population. TBC1 domain family member 25 (TBC1D25) regulates the development of myocardial hypertrophy and cerebral ischemia-reperfusion injury; however, its effect on NAFLD/nonalcoholic steatohepatitis (NASH) has not been reported. In this study, we demonstrated that TBC1D25 expression is upregulated in NASH. TBC1D25 deficiency aggravated hepatic steatosis, inflammation, and fibrosis in NASH. In vitro tests revealed that TBC1D25 overexpression restrained NASH responses. Subsequent mechanistic validation experiments demonstrated that TBC1D25 interfered with NASH progression by inhibiting abnormal lipid accumulation and inflammation. TBC1D25 deficiency significantly promoted NASH occurrence and development. Therefore, TBC1D25 may potentially be used as a clinical therapeutic target for NASH treatment.
Collapse
Affiliation(s)
- Anding Wu
- Department of General Surgery, Huanggang Central Hospital, Huanggang, China
| | - Mao Ye
- Department of Cardiology, HuangGang Central Hospital, Huanggang, China
| | - Tengfei Ma
- Department of Neurology, Huanggang Central Hospital, Huanggang, China
| | - Zhigang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongjie Shi
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Ling Yang
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Maolin Yi
- Surgery of Mammary Gland and Thyroid Gland, Huanggang Central Hospital, Huanggang, China
| | - Huoping Li
- Department of Cardiology, HuangGang Central Hospital, Huanggang, China
| |
Collapse
|
26
|
Recent updates on targeting the molecular mediators of NAFLD. J Mol Med (Berl) 2023; 101:101-124. [PMID: 36792729 DOI: 10.1007/s00109-022-02282-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/29/2022] [Accepted: 12/21/2022] [Indexed: 02/17/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is rapidly becoming the most common disease worldwide in an era of rapid economic growth. NAFLD is a multifactorial disease, involving multiple genetic, metabolic, and environmental factors, and is closely associated with metabolic syndrome, obesity, and cardiovascular disease. NAFLD can be classified into nonalcoholic fatty liver disease (NAFL) and nonalcoholic steatohepatitis (NASH), which can both progress to cirrhosis and even hepatocellular carcinoma (HCC). Due to the enormous burden of NAFLD and its complications, no FDA-approved drugs for the treatment of NAFLD are on the market, and therapeutic targets and drug therapies are being actively investigated. In view of the various pathological mechanisms of NAFLD, numbers of preclinical studies and clinical trials have made rapid progress. This review mainly summarizes the most recently characterized mechanisms and therapeutic targets in each mechanism of NAFLD, focusing on the mechanism and application potential.
Collapse
|
27
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Iron as a therapeutic target in chronic liver disease. World J Gastroenterol 2023; 29:616-655. [PMID: 36742167 PMCID: PMC9896614 DOI: 10.3748/wjg.v29.i4.616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/03/2022] [Accepted: 12/31/2022] [Indexed: 01/20/2023] Open
Abstract
It was clearly realized more than 50 years ago that iron deposition in the liver may be a critical factor in the development and progression of liver disease. The recent clarification of ferroptosis as a specific form of regulated hepatocyte death different from apoptosis and the description of ferritinophagy as a specific variation of autophagy prompted detailed investigations on the association of iron and the liver. In this review, we will present a brief discussion of iron absorption and handling by the liver with emphasis on the role of liver macrophages and the significance of the iron regulators hepcidin, transferrin, and ferritin in iron homeostasis. The regulation of ferroptosis by endogenous and exogenous mod-ulators will be examined. Furthermore, the involvement of iron and ferroptosis in various liver diseases including alcoholic and non-alcoholic liver disease, chronic hepatitis B and C, liver fibrosis, and hepatocellular carcinoma (HCC) will be analyzed. Finally, experimental and clinical results following interventions to reduce iron deposition and the promising manipulation of ferroptosis will be presented. Most liver diseases will be benefited by ferroptosis inhibition using exogenous inhibitors with the notable exception of HCC, where induction of ferroptosis is the desired effect. Current evidence mostly stems from in vitro and in vivo experimental studies and the need for well-designed future clinical trials is warranted.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Liver Research Laboratory, University of Crete Medical School, Heraklion 71003, Greece
| | - Ioannis Tsomidis
- First Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54621, Greece
| | - Argyro Voumvouraki
- First Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54621, Greece
| |
Collapse
|
28
|
Finney AC, Das S, Kumar D, McKinney MP, Cai B, Yurdagul A, Rom O. The interplay between nonalcoholic fatty liver disease and atherosclerotic cardiovascular disease. Front Cardiovasc Med 2023; 10:1116861. [PMID: 37200978 PMCID: PMC10185914 DOI: 10.3389/fcvm.2023.1116861] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/23/2023] [Indexed: 05/20/2023] Open
Abstract
Therapeutic approaches that lower circulating low-density lipoprotein (LDL)-cholesterol significantly reduced the burden of cardiovascular disease over the last decades. However, the persistent rise in the obesity epidemic is beginning to reverse this decline. Alongside obesity, the incidence of nonalcoholic fatty liver disease (NAFLD) has substantially increased in the last three decades. Currently, approximately one third of world population is affected by NAFLD. Notably, the presence of NAFLD and particularly its more severe form, nonalcoholic steatohepatitis (NASH), serves as an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), thus, raising interest in the relationship between these two diseases. Importantly, ASCVD is the major cause of death in patients with NASH independent of traditional risk factors. Nevertheless, the pathophysiology linking NAFLD/NASH with ASCVD remains poorly understood. While dyslipidemia is a common risk factor underlying both diseases, therapies that lower circulating LDL-cholesterol are largely ineffective against NASH. While there are no approved pharmacological therapies for NASH, some of the most advanced drug candidates exacerbate atherogenic dyslipidemia, raising concerns regarding their adverse cardiovascular consequences. In this review, we address current gaps in our understanding of the mechanisms linking NAFLD/NASH and ASCVD, explore strategies to simultaneously model these diseases, evaluate emerging biomarkers that may be useful to diagnose the presence of both diseases, and discuss investigational approaches and ongoing clinical trials that potentially target both diseases.
Collapse
Affiliation(s)
- Alexandra C. Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Sandeep Das
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Dhananjay Kumar
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - M. Peyton McKinney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, United States
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Correspondence: Arif Yurdagul Oren Rom
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Correspondence: Arif Yurdagul Oren Rom
| |
Collapse
|
29
|
Zhou XD, Cai J, Targher G, Byrne CD, Shapiro MD, Sung KC, Somers VK, Chahal CAA, George J, Chen LL, Zhou Y, Zheng MH. Metabolic dysfunction-associated fatty liver disease and implications for cardiovascular risk and disease prevention. Cardiovasc Diabetol 2022; 21:270. [PMID: 36463192 PMCID: PMC9719631 DOI: 10.1186/s12933-022-01697-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/14/2022] [Indexed: 12/07/2022] Open
Abstract
The newly proposed term "metabolic dysfunction-associated fatty liver disease" (MAFLD) is replacing the old term "non-alcoholic fatty liver disease" (NAFLD) in many global regions, because it better reflects the pathophysiology and cardiometabolic implications of this common liver disease. The proposed change in terminology from NAFLD to MAFLD is not simply a single-letter change in an acronym, since MAFLD is defined by a set of specific and positive diagnostic criteria. In particular, the MAFLD definition specifically incorporates within the classification recognized cardiovascular risk factors. Although convincing evidence supports a significant association between both NAFLD and MAFLD, with increased risk of CVD morbidity and mortality, neither NAFLD nor MAFLD have received sufficient attention from the Cardiology community. In fact, there is a paucity of scientific guidelines focusing on this common and burdensome liver disease from cardiovascular professional societies. This Perspective article discusses the rationale and clinical relevance for Cardiologists of the newly proposed MAFLD definition.
Collapse
Affiliation(s)
- Xiao-Dong Zhou
- Department of Cardiovascular Medicine, the Heart Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingjing Cai
- Department of Cardiology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Giovanni Targher
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Verona, Verona, Italy
| | - Christopher D Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton, Southampton General Hospital, Southampton, UK
| | - Michael D Shapiro
- Center for Prevention of Cardiovascular Disease, Section On Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ki-Chul Sung
- Department of Internal Medicine, Division of Cardiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine, Rochester, USA
| | - C Anwar A Chahal
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | - Li-Li Chen
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yong Zhou
- Clinical Research Institute, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China.
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China.
| |
Collapse
|
30
|
Wu X, Huang J, Tang J, Sun Y, Zhao G, Yan C, Liu Z, Yi W, Xu S, Yu X. Isoginkgetin, a bioactive constituent from Ginkgo Biloba, protects against obesity-induced cardiomyopathy via enhancing Nrf2/ARE signaling. Redox Biol 2022; 57:102485. [PMID: 36162256 PMCID: PMC9516449 DOI: 10.1016/j.redox.2022.102485] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/11/2022] [Accepted: 09/19/2022] [Indexed: 11/22/2022] Open
Abstract
Obesity-induced metabolic cardiomyopathy (MC), characterized by lipotoxicity and excessive oxidative stress, emerges as the leading cause of heart failure in the obese patients. Yet, its therapy remains very limited. Here, we demonstrated that isoginkgetin (IGK), a bioactive biflavonoid isolated from medicinal herb Ginkgo Biloba, protected against obesity-induced cardiac diastolic dysfunction and adverse remodeling. Transcriptomics profiling revealed that IGK activated Nrf2 signaling in the heart tissues of the obese mice. Consistent with this observation, IGK treatment increased the nuclear translocation of Nrf2, which in turn trigger the activation of its downstream target genes (e. g. HO-1 and NQO1). In addition, IGK significantly rejuvenated mitochondrial defects in obese heart tissues as evidenced by enhancing mitochondrial respiratory capacity and resisting the collapse of mitochondrial potential and oxidative stress both in vitro and in vivo. Mechanistically, IGK stabilized Nrf2 protein via inhibiting the proteasomal degradation, independent of transcription regulation. Moreover, molecular docking and dynamics simulation assessment demonstrated a good binding mode between IGK and Nrf2/Keap1. Of note, the protective effects conferred by IGK against obesity-induced mitochondrial defects and cardiac dysfunction was compromised by Nrf2 gene silencing both in vitro and in vivo, consolidating a pivotal role of Nrf2 in IGK-elicited myocardial protection against MC. Thus, the present study identifies IGK as a promising drug candidate to alleviate obesity-induced oxidative stress and cardiomyocyte damage through Nrf2 activation, highlighting the therapeutic potential of IGK in ameliorating obesity-induced cardiomyopathy.
Collapse
Affiliation(s)
- Xiaoqian Wu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences& the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511500, China.
| | - Jianrong Huang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences& the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Junyuan Tang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences& the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuling Sun
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences& the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Guojun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511500, China
| | - Cuishi Yan
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences& the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhenghong Liu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230037, China
| | - Wei Yi
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences& the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230037, China.
| | - Xiyong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences& the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
31
|
Xu X, Poulsen KL, Wu L, Liu S, Miyata T, Song Q, Wei Q, Zhao C, Lin C, Yang J. Targeted therapeutics and novel signaling pathways in non-alcohol-associated fatty liver/steatohepatitis (NAFL/NASH). Signal Transduct Target Ther 2022; 7:287. [PMID: 35963848 PMCID: PMC9376100 DOI: 10.1038/s41392-022-01119-3] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/15/2022] [Accepted: 07/08/2022] [Indexed: 11/24/2022] Open
Abstract
Non-alcohol-associated fatty liver/steatohepatitis (NAFL/NASH) has become the leading cause of liver disease worldwide. NASH, an advanced form of NAFL, can be progressive and more susceptible to developing cirrhosis and hepatocellular carcinoma. Currently, lifestyle interventions are the most essential and effective strategies for preventing and controlling NAFL without the development of fibrosis. While there are still limited appropriate drugs specifically to treat NAFL/NASH, growing progress is being seen in elucidating the pathogenesis and identifying therapeutic targets. In this review, we discussed recent developments in etiology and prospective therapeutic targets, as well as pharmacological candidates in pre/clinical trials and patents, with a focus on diabetes, hepatic lipid metabolism, inflammation, and fibrosis. Importantly, growing evidence elucidates that the disruption of the gut-liver axis and microbe-derived metabolites drive the pathogenesis of NAFL/NASH. Extracellular vesicles (EVs) act as a signaling mediator, resulting in lipid accumulation, macrophage and hepatic stellate cell activation, further promoting inflammation and liver fibrosis progression during the development of NAFL/NASH. Targeting gut microbiota or EVs may serve as new strategies for the treatment of NAFL/NASH. Finally, other mechanisms, such as cell therapy and genetic approaches, also have enormous therapeutic potential. Incorporating drugs with different mechanisms and personalized medicine may improve the efficacy to better benefit patients with NAFL/NASH.
Collapse
Affiliation(s)
- Xiaohan Xu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Kyle L Poulsen
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Lijuan Wu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shan Liu
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Tatsunori Miyata
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Qiaoling Song
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Qingda Wei
- School of Medicine, Zhengzhou University, Zhengzhou, China
| | - Chenyang Zhao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Chunhua Lin
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jinbo Yang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
32
|
Impact of NAFLD and its pharmacotherapy on lipid profile and CVD. Atherosclerosis 2022; 355:30-44. [PMID: 35872444 DOI: 10.1016/j.atherosclerosis.2022.07.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/16/2022] [Accepted: 07/13/2022] [Indexed: 11/21/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of death worldwide. Increasing evidence suggests that, in addition to traditional metabolic risk factors such as obesity, hypercholesterolemia, hypertension, diabetes mellitus, and insulin resistance (IR), nonalcoholic fatty liver disease (NAFLD) is an emerging driver of ASCVD via multiple mechanisms, mainly by disrupting lipid metabolism. The lack of pharmaceutical treatment has spurred substantial investment in the research and development of NAFLD drugs. However, many reagents with promising therapeutic potential for NAFLD also have considerable impacts on the circulating lipid profile. In this review, we first summarize the mechanisms linking lipid dysregulation in NAFLD to the progression of ASCVD. Importantly, we highlight the potential risks of/benefits to ASCVD conferred by NAFLD pharmaceutical treatments and discuss potential strategies and next-generation drugs for treating NAFLD without the unwanted side effects.
Collapse
|
33
|
Deng KQ, Huang X, Lei F, Zhang XJ, Zhang P, She ZG, Cai J, Ji YX, Li H. Role of hepatic lipid species in the progression of nonalcoholic fatty liver disease. Am J Physiol Cell Physiol 2022; 323:C630-C639. [PMID: 35759443 DOI: 10.1152/ajpcell.00123.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most common liver disease due to the global pandemic of metabolic diseases. Dysregulation of hepatic lipid metabolism plays a central role in the initiation and progression of NAFLD. With the advancement of lipidomics, an increasing number of lipid species and underlying mechanisms associating hepatic lipid components have been revealed. Therefore, the focus of this mini-review is to highlight the links between hepatic lipid species and their mechanisms mediating the pathogenesis of NAFLD. We first summarized the interplay between NAFLD and hepatic lipid disturbances. Next, we focused on reviewing the role of saturated fatty acids, cholesterol, oxidized phospholipids, and their respective intermediates in the pathogenesis of NAFLD. The mechanisms by which monounsaturated fatty acids and other pro-resolving mediators exert protective effects are also addressed. Finally, we further discussed the implication of different analysis approaches in lipidomic. Evolving insights into the pathophysiology of NAFLD will provide the opportunity for drug development.
Collapse
Affiliation(s)
- Ke-Qiong Deng
- Department of Cardiology, Center Hospital of Huanggang, Huanggang, China.,Huanggang Institute of Translation Medicine, Huanggang, China.,Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuewei Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animal, Wuhan University, Wuhan, China
| | - Fang Lei
- Institute of Model Animal, Wuhan University, Wuhan, China.,School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China.,School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Peng Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China.,School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animal, Wuhan University, Wuhan, China
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan-Xiao Ji
- Institute of Model Animal, Wuhan University, Wuhan, China.,School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Hongliang Li
- Huanggang Institute of Translation Medicine, Huanggang, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animal, Wuhan University, Wuhan, China
| |
Collapse
|
34
|
Bian H, Liu YM, Chen ZN. New avenues for NASH therapy by targeting ACC. Cell Metab 2022; 34:191-193. [PMID: 35108509 DOI: 10.1016/j.cmet.2022.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Acetyl-CoA carboxylase (ACC) is one of the more promising therapeutic targets for non-alcoholic steatohepatitis (NASH), but current ACC inhibitors already tested in clinical trials also exert the unwanted adverse side effect of hypertriglyceridemia. In two recent studies by Calle et al. in Nature Medicine and Zhang et al. in Science Translational Medicine, new strategies for ACC targeting were explored for NASH therapy that successfully resolved the adverse effect of hyperlipidemia while maintaining potent anti-NASH efficacy. These findings bring encouraging new momentum to the clinical application of ACC inhibition for NASH therapy.
Collapse
Affiliation(s)
- Huijie Bian
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Ye-Mao Liu
- Department of Cardiology, Huanggang Central Hospital, Huanggang, China; Huanggang Institute of Translational Medicine, Huanggang, China
| | - Zhi-Nan Chen
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
35
|
The multifaceted role of ferroptosis in liver disease. Cell Death Differ 2022; 29:467-480. [PMID: 35075250 PMCID: PMC8901678 DOI: 10.1038/s41418-022-00941-0] [Citation(s) in RCA: 362] [Impact Index Per Article: 120.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
Ferroptosis is an iron-dependent form of non-apoptotic cell death characterized by excessive lipid peroxidation and associated with a plethora of pathological conditions in the liver. Emerging evidence supports the notion that dysregulated metabolic pathways and impaired iron homeostasis play a role in the progression of liver disease via ferroptosis. Although the molecular mechanisms by which ferroptosis causes disease are poorly understood, several ferroptosis-associated genes and pathways have been implicated in liver disease. Here, we review the physiological role of the liver in processing nutrients, our current understanding of iron metabolism, the characteristics of ferroptosis, and the mechanisms that regulate ferroptosis. In addition, we summarize the role of ferroptosis in the pathogenesis of liver disease, including liver injury, non-alcoholic steatohepatitis, liver fibrosis, liver cirrhosis, and hepatocellular carcinoma. Finally, we discuss the therapeutic potential of targeting ferroptosis for managing liver disease.
Collapse
|
36
|
Zhang XJ, She ZG, Wang J, Sun D, Shen LJ, Xiang H, Cheng X, Ji YX, Huang YP, Li PL, Yang X, Cheng Y, Ma JP, Wang HP, Hu Y, Hu F, Tian S, Tian H, Zhang P, Zhao GN, Wang L, Hu ML, Yang Q, Zhu LH, Cai J, Yang J, Zhang X, Ma X, Xu Q, Touyz RM, Liu PP, Loomba R, Wang Y, Li H. Multiple omics study identifies an interspecies conserved driver for nonalcoholic steatohepatitis. Sci Transl Med 2021; 13:eabg8117. [PMID: 34910546 DOI: 10.1126/scitranslmed.abg8117] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Junyong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Dating Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Li-Jun Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hui Xiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xu Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yan-Xiao Ji
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yong-Ping Huang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Peng-Long Li
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xia Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yanjie Cheng
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jun-Peng Ma
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Hai-Ping Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yufeng Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Fengjiao Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Han Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Peng Zhang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Guang-Nian Zhao
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Lin Wang
- Department of Hepatic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Man-Li Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qin Yang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Li-Hua Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Juan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xin Zhang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19004, USA
| | - Qingbo Xu
- Centre for Clinic Pharmacology, The William Harvey Research Institute, Queen Mary University of London, London SE5 9NU, UK
| | - Rhian M Touyz
- British Heart Foundation Chair in Cardiovascular Medicine, and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Peter P Liu
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Epidemiology, University of California, San Diego, San Diego, CA 92093, USA
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Institute of Model Animal of Wuhan University, Wuhan 430071, China.,School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
37
|
Using chimeric antigen receptor T-cell therapy to fight glioblastoma multiforme: past, present and future developments. J Neurooncol 2021; 156:81-96. [PMID: 34825292 PMCID: PMC8714623 DOI: 10.1007/s11060-021-03902-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022]
Abstract
Introduction Glioblastoma multiforme (GBM) constitutes one of the deadliest tumors to afflict humans, although it is still considered an orphan disease. Despite testing multiple new and innovative therapies in ongoing clinical trials, the median survival for this type of malignancy is less than two years after initial diagnosis, regardless of therapy. One class of promising new therapies are chimeric antigen receptor T cells or CAR-T which have been shown to be very effective at treating refractory liquid tumors such as B-cell malignancies. However, CAR-T effectivity against solid tumors such as GBM has been limited thus far. Methods A Pubmed, Google Scholar, Directory of Open Access Journals, and Web of Science literature search using the terms chimeric antigen receptor or CAR-T, GBM, solid tumor immunotherapy, immunotherapy, and CAR-T combination was performed for publication dates between January 1987 and November 2021. Results In the current review, we present a comprehensive list of CAR-T cells developed to treat GBM, we describe new possible T-cell engineering strategies against GBM while presenting a short introductory history to the reader regarding the origin(s) of this cutting-edge therapy. We have also compiled a unique list of anti-GBM CAR-Ts with their specific protein sequences and their functions as well as an inventory of clinical trials involving CAR-T and GBM. Conclusions The aim of this review is to introduce the reader to the field of T-cell engineering using CAR-Ts to treat GBM and describe the obstacles that may need to be addressed in order to significantly delay the relentless growth of GBM. Supplementary Information The online version contains supplementary material available at 10.1007/s11060-021-03902-8.
Collapse
|