1
|
Liu D, Wang C, Chen Y, Huang X, Wen Y, Duan S, Cai Y, Li X, He J, Han K, Li T, Li Y, Xia Z. Protein Kinase C Epsilon Overexpression Protects the Heart Against Doxorubicin-Induced Cardiotoxicity Via Activating SIRT1. Cardiovasc Toxicol 2025; 25:915-928. [PMID: 40327286 PMCID: PMC12116906 DOI: 10.1007/s12012-025-09995-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 04/05/2025] [Indexed: 05/07/2025]
Abstract
Doxorubicin (DOX)-induced cardiotoxicity (DIC) is known to be associated with reduction of cardiac protein kinase C epsilon (PKC-ε). PKC-ε promotes cell survival and protects hearts against various stresses. However, it is unclear whether or not the reduction in cardiac PKC-ε expression plays a causal role in DIC and in particular the potential underlying mechanism whereby PKC-ε may protect against DIC. C57BL/6 mice (8-10-week-old) were either treated with DOX administered intraperitoneally for a duration of 4 weeks to produce cardiotoxicity, or untreated in which mice received the same volume of saline. In vitro, neonatal rat ventricle cardiomyocytes were exposed to DOX for 24 h in the absence or presence of adenovirus overexpressing PKC-ε. Cardiomyocytes in a subgroup were treated with sirtuin-1 (SIRT1) selective inhibitor Ex527. Four weeks after DOX, cardiac contractile function was decreased concomitant with increased serum CK-MB and LDH levels as well as increases in Bax-to-Bcl-2 ratio and Cleaved Caspase 3 proteins expression, while PKC-ε and Sirt1 protein expressions were significantly decreased. In vitro, DOX reduced cardiomyocyte PKC-ε and SIRT1 protein expression, decreased cardiomyocyte viability, and increased LDH release with concomitant increases in oxidative stress and apoptosis. These changes were attenuated by overexpression of PKC-ε. IP study showed that PKC-ε could directly or indirectly bind SIRT1 in cardiomyocytes, and the protect effects of PKC-ε were further canceled by SIRT1 inhibition. In conclusion, activating SIRT1 may represent a major mechanism whereby PKC-ε protects the heart against DOX-induced cell apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Danyong Liu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Chunyan Wang
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Yao Chen
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Xiaolei Huang
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Yajie Wen
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Shan Duan
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China
| | - Yin Cai
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, China
| | - Xia Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jianfeng He
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Kaijia Han
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Ting Li
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Yuantao Li
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518038, Guangdong, China.
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Pok Fu Lam Road, Hong Kong, China.
- Doctoral Training Platform for Research and Translation, BoShiWan, GuanChong Village, Shuanghe Town, ZhongXiang, 431913, Hubei, China.
| |
Collapse
|
2
|
Gao T, Tang Y, Zeng T, Wang J, Zhang X, Liu Q, Guan X, Tang X, Lu G, Li J, Liu M, Zhang D, Lv S, Gu J. Neuraminidase 1 Exacerbated Glycolytic Dysregulation and Cardiotoxicity by Destabilizing SIRT1 through Interactions with NRF2 and HIF1α. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2414504. [PMID: 40411250 DOI: 10.1002/advs.202414504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/20/2025] [Indexed: 05/26/2025]
Abstract
Despite significant therapeutic advances, cumulative DOX-induced cardiotoxicity (DIC) events remain unacceptably high. Recent evidence has underscored the critical role of impaired glycolytic metabolism in cardiovascular damage. Neuraminidase 1 (NEU1), a member of the neuraminidase family, catalyzes the hydrolysis of terminal sialic acids from glycoconjugates. Here, it is aimed to characterize the role of NEU1 on defective glycolysis during DIC. Mouse models with cardiac-specific genetic modifications of Neu1, Nrf2, and Sirt1 underwent functional analyses, and RNA sequencing to clarify NEU1's role in glycolytic metabolism during DIC. It is discovered that NEU1 is highly expressed after DOX exposure and positively correlated with defective glycolysis phenotypes. Cardiomyocyte-specific deficiency of Neu1 ameliorated impaired glycolytic metabolism and DIC, whereas overexpression of Neu1 in cardiomyocytes exacerbated these pathological phenotypes. Mechanistically, the upregulation of Neu1 is attributed to HIF1α's transcriptional repression, which necessitated the collaboration of NRF2. Additionally, the C-terminal region of NEU1 physically interacted with SIRT1, facilitating its lysosomal-mediated degradation and contributing to the aberrant glycolytic phenotype. The pharmacological or genetic manipulation of NRF2 and HIF1α remarkably abolished DOX-induced NEU1 upregulation, compromised glucose metabolism, and DIC progression. Collectively, NEU1 as a key regulator of cardiac glycolysis is established, offering new therapeutic avenues for DIC through maintaining metabolic flexibility.
Collapse
Affiliation(s)
- Ting Gao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, China
| | - Tao Zeng
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaohui Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Qingbo Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xun Guan
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xinyu Tang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jiahao Li
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Mingrui Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Dongmei Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Sixuan Lv
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
3
|
Wang Y, Chen Y, Yang J, Sun W, Zhang X. Electro-Acupuncture Therapy Alleviates Post-Stroke Insomnia by Regulating Sirt1 and the Nrf2-ARE Pathway. Neuromolecular Med 2025; 27:37. [PMID: 40381125 DOI: 10.1007/s12017-025-08862-0] [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: 03/13/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
Post-stroke insomnia (PSI) is a common complication following stroke, which seriously affects patients' life quality. Electro-acupuncture (EA) is an innovative form of traditional Chinese acupuncture that combines electricity with needles to achieve the prevention and treatment of diseases. However, there is limited understanding regarding the treatment mechanism of EA in PSI. In our study, we aimed to investigate the role of EA on PSI development. Our study findings indicated that the quality of sleep, levels of neurotransmitters 5-hydroxytryptamine (5-HT) and gamma-aminobutyric acid (γ-GABA), and antioxidant levels showed significant improvement following EA treatment in PSI clinical samples and rat models, while the levels of pro-inflammatory factor interleukin 6 (IL-6), tumor necrosis factor-alpha (TNF-α), and astrocyte damage were notably reduced. Furthermore, it was discovered that the levels of sirtuin 1 (Sirt1) were reduced in PSI, a condition that was significantly ameliorated by EA treatment. Additionally, the inhibition of Sirt1 caused a marked elevation in astrocyte apoptosis, inflammatory response, and oxidative stress. Besides, the nuclear factor E2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway was deactivated in the PSI rat model and Sirt1-silenced cells. However, the suppressive impact was successfully counteracted by EA or estazolam (ES), and the overexpression of Nrf2 partially alleviated the increase in apoptosis, inflammation, and oxidative stress caused by Sirt1 knockdown. Taken together, these findings indicated that EA improved sleep quality and silenced Sirt1-induced apoptosis, inflammation, and oxidative stress in PSI by activating the Nrf2-ARE pathway.
Collapse
Affiliation(s)
- Yiming Wang
- The Fourth Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region Chinese Medicine Hospital, Urumqi, Xinjiang, China
| | - Yifei Chen
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jianbo Yang
- Second Department of Neurology, The Fourth Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region Chinese Medicine Hospital, No.116, Huanghe Road, Shaybak District, Urumqi, 830000, Xinjiang, China
| | - Wei Sun
- Second Department of Neurology, The Fourth Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region Chinese Medicine Hospital, No.116, Huanghe Road, Shaybak District, Urumqi, 830000, Xinjiang, China
| | - Xiaoning Zhang
- Second Department of Neurology, The Fourth Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region Chinese Medicine Hospital, No.116, Huanghe Road, Shaybak District, Urumqi, 830000, Xinjiang, China.
| |
Collapse
|
4
|
Ma Y, Wang Y, Chen R, Wang Y, Fang Y, Qin C, Wang T, Shen X, Zhou T, Tian L, Sun T, Fan L, Wang X, Han D, Cao F. Exosomal transfer of pro-pyroptotic miR-216a-5p exacerbates anthracycline cardiotoxicity through breast cancer-heart pathological crosstalk. Signal Transduct Target Ther 2025; 10:157. [PMID: 40360476 PMCID: PMC12075849 DOI: 10.1038/s41392-025-02245-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/26/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
Doxorubicin (DOX) is the most effective chemotherapeutic for breast cancer, but it is usually associated with severe cardiotoxicity. Further investigation to alleviate its side effects is essential. The present study investigated the mechanism of the cross-organ communication between tumors and the heart and potential intervention targets. Morphological bubble-like protrusions were observed in both adult murine ventricular cardiomyocytes (AMVCs) and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) cocultured with breast cancer cells (BCCs), along with elevated expression of pyroptosis-related proteins. Exosomes (EXOs) from DOX-treated BCCs aggravated DOX-induced cardiotoxicity (DOXIC) in an orthotopic mouse model of breast cancer. Blocking miRNAs by knocking down Rab27a or inhibiting the release of EXOs in cancer tissue by Dicer enzyme knockout attenuated this additional injury effect. Exosomal miRNA sequencing revealed that miR-216a-5p is especially upregulated in EXOs from DOX-induced BCCs. Mechanistically, miR-216a-5p was upregulated by enhanced transcription mediated by DOX-induced AMP-dependent transcription factor 3 (ATF3) and packaged into EXOs by splicing factor 3b subunit 4 (SF3B4) in BCCs. Itchy E3 ubiquitin-protein ligase (ITCH) was identified as a novel downstream target mRNA of miR-216a-5p. ITCH negatively mediated thioredoxin-interacting protein (TXNIP) ubiquitination to activate the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome pathway, ultimately leading to cardiomyocyte pyroptosis. Our findings revealed novel cross-organ pathogenic communication between breast cancer and the heart through the exosomal miR-216a-5p-mediated ITCH/TXNIP/NLRP3 pathway, which drives cardiomyocyte pyroptosis. These findings suggest that targeting myocardial miR-216a-5p or blocking harmful EXOs from breast cancer is a potential therapeutic strategy for alleviating DOXIC.
Collapse
Affiliation(s)
- Yan Ma
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Yongjun Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Renzheng Chen
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Yabin Wang
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Yan Fang
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Cheng Qin
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Tianhu Wang
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Xiaoying Shen
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Tingwen Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Lei Tian
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Ting Sun
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Li Fan
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Xiaoning Wang
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China.
- School of Life Sciences, Fudan University, Shanghai, China. Room 1910, West Guanghua Tower, 220 Handan Road, Shanghai, 200433, China.
| | - Dong Han
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China.
| | - Feng Cao
- Chinese Military Medical School, National Clinical Research Center for Geriatric Diseases, The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China.
| |
Collapse
|
5
|
Xiong X, Huang H, Wang N, Zhou K, Song X. Sirt1 overexpression inhibits chondrocyte ferroptosis via Ftl deacetylation to suppress the development of osteoarthritis. J Bone Miner Metab 2025; 43:203-215. [PMID: 39786573 DOI: 10.1007/s00774-024-01574-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/09/2024] [Indexed: 01/12/2025]
Abstract
INTRODUCTION Osteoarthritis (OA) is a chronic degenerative joint disorder characterized by an imbalance in chondrocyte metabolism. Ferroptosis has been implicated in the pathogenesis of OA. The role of Sirt1, a deacetylase, in mediating deacetylation during ferroptosis in OA chondrocytes remains underexplored. This study aimed to elucidate the mechanisms by which Sirt1 influences chondrocyte ferroptosis in the development of OA. MATERIALS AND METHODS In vitro and in vivo models of OA were established using IL-1β-induced mouse chondrocytes and a destabilization of the medial meniscus (DMM) mouse model, respectively. Ferroptosis was evaluated through measurements of cell viability, lactate dehydrogenase (LDH) release, intracellular levels of Fe2+, glutathione (GSH), malondialdehyde (MDA), lipid reactive oxygen species (ROS), propidium iodide staining, and Western blot analysis. The underlying mechanisms were further investigated using quantitative real-time polymerase chain reaction, Western blotting, immunoprecipitation (IP), co-immunoprecipitation (Co-IP), and glutathione-S-transferase pulldown assays. In vivo validation was performed via Safranin O staining. RESULTS IL-1β induced ferroptosis and increased histone acetylation, effects that were partially reversed by Sirt1 overexpression. Mechanistically, Sirt1 overexpression upregulated ferritin light polypeptide (Ftl) expression by deacetylating Ftl at the K181 residue. Ftl knockdown inhibited the ferroptosis-enhancing effect of Sirt1 overexpression in chondrocytes. In vivo studies showed that Sirt1 overexpression mitigated the progression of OA and reduced ferroptosis in the DMM-induced OA mouse model. CONCLUSION Our findings confirm that Sirt1 overexpression promotes Ftl expression through deacetylation at the K181 site, thereby suppressing chondrocyte ferroptosis and attenuating the progression of OA. These results suggest a potential therapeutic target for OA treatment.
Collapse
Affiliation(s)
- Xiaolong Xiong
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Hui Huang
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ning Wang
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Kai Zhou
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xinghui Song
- Universiti Kebangsaan Malaysia Health Science, UKM, 43600, Bandar Baru Bangi, Selangor, Malaysia.
| |
Collapse
|
6
|
Muttiah B, Hanafiah A. Gut Microbiota and Cardiovascular Diseases: Unraveling the Role of Dysbiosis and Microbial Metabolites. Int J Mol Sci 2025; 26:4264. [PMID: 40362500 PMCID: PMC12072866 DOI: 10.3390/ijms26094264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiovascular diseases (CVDs), including heart failure (HF), hypertension, myocardial infarction (MI), and atherosclerosis, are increasingly linked to gut microbiota dysbiosis and its metabolic byproducts. HF, affecting over 64 million individuals globally, is associated with systemic inflammation and gut barrier dysfunction, exacerbating disease progression. Similarly, hypertension and MI correlate with reduced microbial diversity and an abundance of pro-inflammatory bacteria, contributing to vascular inflammation and increased cardiovascular risk. Atherosclerosis is also influenced by gut dysbiosis, with key microbial metabolites such as trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFAs) playing crucial roles in disease pathogenesis. Emerging evidence highlights the therapeutic potential of natural compounds, including flavonoids, omega-3 fatty acids, resveratrol, curcumin, and marine-derived bioactives, which modulate the gut microbiota and confer cardioprotective effects. These insights underscore the gut microbiota as a critical regulator of cardiovascular health, suggesting that targeting dysbiosis may offer novel preventive and therapeutic strategies. Further research is needed to elucidate underlying mechanisms and optimize microbiome-based interventions for improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Barathan Muttiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Alfizah Hanafiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- GUT Research Group, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
7
|
Luo Y, Wu H, Min X, Chen Y, Deng W, Chen M, Yang C, Xiong H. SIRT1 prevents noise-induced hearing loss by enhancing cochlear mitochondrial function. Cell Commun Signal 2025; 23:160. [PMID: 40176044 PMCID: PMC11963675 DOI: 10.1186/s12964-025-02152-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/11/2025] [Indexed: 04/04/2025] Open
Abstract
Exposure to traumatic noise triggers cochlear damage and consequently causes permanent sensorineural hearing loss. However, effective treatment strategies for noise-induced hearing loss (NIHL) are lacking. Sirtuin 1 (SIRT1) is a NAD+-dependent deacetylase that plays a critical role in multiple physiological and pathological events. However, its role in NIHL pathogenesis remains elusive. This study revealed that SIRT1 expression in the cochlea progressively decreases in a mouse model of NIHL. Hair cell-specific knockout of SIRT1 exacerbates the noise-induced loss of outer and inner hair cell synaptic ribbons, retraction of cochlear nerve terminals, and oxidative stress, leading to more severe NIHL. Conversely, adeno-associated virus (AAV)-mediated SIRT1 overexpression effectively attenuated most noise-induced cochlear damage and alleviated NIHL. Transcriptomic analysis revealed that SIRT1 deficiency impairs glucose metabolism and inhibits antioxidant pathways in the cochlea following exposure to noise. Further investigation revealed that SIRT1 exerts an antioxidant effect, at least in part, through AMPK activation in cultured auditory HEI-OC1 cells exposed to oxidative stress. Collectively, these findings indicate that SIRT1 is essential for the maintenance of redox balance and mitochondrial function in the cochlea after traumatic noise exposure, thus providing a promising therapeutic target for NIHL treatment.
Collapse
Affiliation(s)
- Yuelian Luo
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Haoyang Wu
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xin Min
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yi Chen
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wenting Deng
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Minjun Chen
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chuxuan Yang
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hao Xiong
- Department of Otolaryngology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Hong L, Cai X, Zhan Y, Liu S, Zou P, Chen Y, Shao L. TLR2 activates AP-1 to facilitate CTGF transcription and stimulate doxorubicin-induced myocardial injury. Br J Pharmacol 2025. [PMID: 40097259 DOI: 10.1111/bph.17423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/17/2024] [Accepted: 11/11/2024] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND AND PURPOSE Our study aimed to explore the mechanistic network of toll-like receptor 2 (TLR2)/activator protein-1 (AP-1) combined with SOX10 activation of the mitogen-activated protein kinase (MAPK) pathway via connective tissue growth factor (CTGF) in doxorubicin (Dox)-induced myocardial injury. EXPERIMENTAL APPROACH Rats with Dox-induced myocardial injury were treated with a TLR2 inhibitor or CTGF silencing lentiviral vector. H9c2 cells were treated with genetic vectors or MAPK pathway activators. Cardiac function was tested using echocardiography and serum markers. H&E, Sirius red and TUNEL staining were used to detect myocardial pathological changes, collagen accumulation and apoptosis. Western blot was used to detect proteins related to cardiac hypertrophy, fibrosis, apoptosis and the MAPK pathway. H9c2 cell injury was assessed by testing cell viability, lactate dehydrogenase (LDH) release and mitochondrial membrane potential. KEY RESULTS TLR2 and CTGF were highly expressed in patients with heart failure, and Dox treatment further increased their expression. Inhibiting TLR2 or silencing CTGF improved cardiac function and reduced myocardial fibrosis and apoptosis in Dox-treated rats. Silencing of TLR2 alleviated Dox-induced H9c2 cell injury, which was nullified by CTGF overexpression. TLR2 activated AP-1, which cooperated with SOX10 to promote CTGF transcription. MAPK activation aggravated H9c2 cells against Dox-induced injury. CONCLUSIONS AND IMPLICATIONS TLR2 activates AP-1 which cooperates with SOX10 to promote CTGF transcription and subsequently activate the MAPK pathway, thereby stimulating Dox-induced myocardial injury.
Collapse
Affiliation(s)
- Lang Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xinyong Cai
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yuliang Zhan
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Songtao Liu
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Pengtao Zou
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yanmei Chen
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Liang Shao
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
9
|
Guo Y, Wang J, Zhang D, Tang Y, Cheng Q, Li J, Gao T, Zhang X, Lu G, Liu M, Guan X, Tang X, Gu J. Diabetes-associated sleep fragmentation impairs liver and heart function via SIRT1-dependent epigenetic modulation of NADPH oxidase 4. Acta Pharm Sin B 2025; 15:1480-1496. [PMID: 40370565 PMCID: PMC12069238 DOI: 10.1016/j.apsb.2024.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 05/16/2025] Open
Abstract
Although clinical evidence suggests that nonalcoholic fatty liver disease is an established major risk factor for heart failure, it remains unexplored whether sleep disorder-caused hepatic damage contributes to the development of cardiovascular disease (CVD). Here, our findings revealed that sleep fragmentation (SF) displayed notable hepatic detrimental phenotypes, including steatosis and oxidative damage, along with significant abnormalities in cardiac structure and function. All these pathological changes persisted even after sleep recovery for 2 consecutive weeks or more, displaying memory properties. Mechanistically, persistent higher expression of nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) in the liver was the key initiator of SF-accelerated damage phenotypes. SF epigenetically controlled the acetylation of histone H3 lysine 27 (H3K27ac) enrichment at the Nox4 promoter and markedly increased Nox4 expression in liver even after sleep recovery. Moreover, fine coordination of the circadian clock and hepatic damage was strictly controlled by BMAL1-dependent Sirtuin 1 (Sirt1) transcription after circadian misalignment. Accordingly, genetic manipulation of liver-specific Nox4 or Sirt1, along with pharmacological intervention targeting NOX4 (GLX351322) or SIRT1 (Resveratrol), could effectively erase the epigenetic modification of Nox4 by reducing the H3K27ac level and ameliorate the progression of liver pathology, thereby counteracting SF-evoked sustained CVD. Collectively, our findings may pave the way for strategies to mitigate myocardial injury from persistent hepatic detrimental memory in diabetic patients.
Collapse
Affiliation(s)
- Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dongmei Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, the First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Quanli Cheng
- Department of Cardiovascular Disease, First Hospital of Jilin University, Changchun 130021, China
| | - Jiahao Li
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Ting Gao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaohui Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Mingrui Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xun Guan
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinyu Tang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
10
|
Abukhalil MH, Al-Alami Z, Alfwuaires MA, Imran MR, Aladaileh SH, Althunibat OY. Taxifolin Protects Against 5-Fluorouracil-Induced Cardiotoxicity in Mice Through Mitigating Oxidative Stress, Inflammation, and Apoptosis: Possible Involvement of Sirt1/Nrf2/HO-1 Signaling. Cardiovasc Toxicol 2025; 25:455-470. [PMID: 39827225 DOI: 10.1007/s12012-025-09962-w] [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: 10/09/2024] [Accepted: 01/12/2025] [Indexed: 01/22/2025]
Abstract
Although 5-fluorouracil (5-FU) is widely utilized in cancer treatment, its side effects, including cardiotoxicity, limit its use. Taxifolin (TAX) is a bioactive anti-inflammatory and antioxidant flavonoid. This study aimed to elucidate the protective effect of TAX against 5-FU-induced cardiac injury in male mice. Mice were treated with TAX (25 and 50 mg/kg, orally) for 10 days and a single dose of 150 mg/kg 5-FU at day 8. Mice intoxicated with 5-FU showed increased creatine kinase-MB and lactate dehydrogenase activities and troponin I levels, with multiple cardiac histopathological changes. They also showed a significant increase in cardiac malondialdehyde (MDA) and nitric oxide (NO) and decreases in myocardial reduced glutathione (GSH) content and superoxide dismutase (SOD) and catalase (CAT) activities (P < 0.001). Pretreatment of 5-FU-injected mice with TAX suppressed cardiac injury, decreased MDA and NO contents (P < 0.001), and boosted antioxidant defenses in the myocardium. Moreover, TAX attenuated cardiac inflammatory response, as evidenced by the decreased expression levels of cardiac NF-κB p65, inducible nitric oxide synthase (iNOS), and pro-inflammatory cytokines (P < 0.001). Largely, TAX ameliorated the decrease in Bcl-2 expression and the increase in BAX and caspase-3 in the heart. It also restored the cardiac Sirt1/Nrf2/HO-1 signaling pathway. In conclusion, TAX showed significant cardioprotective effects on 5-FU-induced cardiac injury and might represent a promising adjuvant in preventing cardiac injury associated with oxidative stress and inflammation.
Collapse
Affiliation(s)
- Mohammad H Abukhalil
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma'an, 71111, Jordan.
- Department of Biology, College of Science, Al-Hussein Bin Talal University, Ma'an, 71111, Jordan.
| | - Zina Al-Alami
- Department of Basic Medical Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Manal A Alfwuaires
- Department of Biological Sciences, Faculty of Science, King Faisal University, 31982, Al-Ahsa, Saudi Arabia
| | - Mohd Rasheeduddin Imran
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al Batin, 39553, Hafr Al Batin, Saudi Arabia
| | - Saleem H Aladaileh
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al Batin, 39553, Hafr Al Batin, Saudi Arabia
| | - Osama Y Althunibat
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma'an, 71111, Jordan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Jadara University, Irbid, 21110, Jordan
| |
Collapse
|
11
|
Ding YN, Wang HY, Chen XF, Tang X, Chen HZ. Roles of Sirtuins in Cardiovascular Diseases: Mechanisms and Therapeutics. Circ Res 2025; 136:524-550. [PMID: 40014680 DOI: 10.1161/circresaha.124.325440] [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] [Indexed: 03/01/2025]
Abstract
Cardiovascular diseases (CVDs) are experiencing a rapid surge and are widely recognized as the leading cause of mortality in the current aging society. Given the multifactorial etiology of CVDs, understanding the intricate molecular and cellular mechanisms is imperative. Over the past 2 decades, many scientists have focused on Sirtuins, a family of nicotinamide adenine dinucleotide-dependent deacylases. Sirtuins are highly conserved across species, from yeasts to primates, and play a crucial role in linking aging and diseases. Sirtuins participate in nearly all key physiological and pathological processes, ranging from embryogenic development to stress response and aging. Abnormal expression and activity of Sirtuins exist in many aging-related diseases, while their activation has shown efficacy in mitigating these diseases (eg, CVDs). In terms of research, this field has maintained fast, sustained growth in recent years, from fundamental studies to clinical trials. In this review, we present a comprehensive, up-to-date discussion on the biological functions of Sirtuins and their roles in regulating cardiovascular biology and CVDs. Furthermore, we highlight the latest advancements in utilizing Sirtuin-activating compounds and nicotinamide adenine dinucleotide boosters as potential pharmacological targets for preventing and treating CVDs. The key unresolved issues in the field-from the chemicobiological regulation of Sirtuins to Sirtuin-targeted CVD investigations-are also discussed. This timely review could be critical in understanding the updated knowledge of Sirtuin biology in CVDs and facilitating the clinical accessibility of Sirtuin-targeting interventions.
Collapse
Affiliation(s)
- Yang-Nan Ding
- Department of Laboratory Medicine, Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, The Third Affiliated Hospital of Zhengzhou University, China (Y.-N.D.)
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.-N.D., H.-Y.W., H.-Z.C.)
| | - Hui-Yu Wang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.-N.D., H.-Y.W., H.-Z.C.)
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing (H.-Y.W., H.-Z.C.)
| | - Xiao-Feng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, China (X.-F.C.)
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu (X.T.)
| | - Hou-Zao Chen
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.-N.D., H.-Y.W., H.-Z.C.)
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing (H.-Y.W., H.-Z.C.)
| |
Collapse
|
12
|
Tang D, Jin H, Lin M, Jiang F, Wu J. Leonurine alleviates doxorubicin-induced myocarditis in mice via MAPK/ERK pathway inhibition. Am J Transl Res 2025; 17:806-817. [PMID: 40092103 PMCID: PMC11909527 DOI: 10.62347/kwrf2732] [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: 09/02/2024] [Accepted: 12/20/2024] [Indexed: 03/19/2025]
Abstract
OBJECTIVE To investigate the effects of naturally derived leonurine (Leo) on doxorubicin (Dox)-induced myocarditis and analyze its potential mechanisms. METHODS Dox was intraperitoneally injected to establish a myocardial injury model in mice. The effect of Leo on inflammatory cytokine levels in myocardial tissue was assessed by ELISA. Pathological changes in myocardial tissue and apoptosis in myocardial cells were observed using hematoxylin-eosin (HE) and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining. Protein levels were analyzed by Western blot (WB). Mouse myocardial H9c2 cells were divided into control group, Dox group, Leo (10 μmol/L) + Dox group, and Leo (20 μmol/L) + Dox group. Cell viability was assessed using Cell Counting Kit-8 (CCK8), and the levels of inflammatory cytokines were measured. The oxidation level and protein levels in H9c2 cells were also detected. RESULTS Leo significantly reduced the levels of inflammatory cytokines in both serum and cell culture supernatant. Additionally, Leo also decreased the levels of inflammatory cytokines in cardiac tissue. Moreover, Leo suppressed Dox-induced myocardial cell apoptosis by modulating the BCL2 signaling pathway. In vitro studies revealed that both inflammatory cytokines and oxidative stress markers were decreased after treatment with Leo. CONCLUSION Leo exerts significant cardioprotective effects through anti-inflammatory mechanisms, likely mitigating Dox-induced myocardial inflammation by inhibiting the activation of MAPK/ERK pathways. These findings highlight Leo's potential as a promising cardioprotective agent, underscoring its therapeutic promise.
Collapse
Affiliation(s)
- Dachao Tang
- Department of Cardiovascular Medicine, Wenzhou Hospital of Traditional Chinese Medicine Wenzhou 325000, Zhejiang, China
| | - Hu Jin
- Department of Cardiovascular Medicine, Wenzhou Hospital of Traditional Chinese Medicine Wenzhou 325000, Zhejiang, China
| | - Meise Lin
- Department of Cardiovascular Medicine, Wenzhou Hospital of Traditional Chinese Medicine Wenzhou 325000, Zhejiang, China
| | - Fuling Jiang
- Department of Cardiovascular Medicine, Wenzhou Hospital of Traditional Chinese Medicine Wenzhou 325000, Zhejiang, China
| | - Jing Wu
- Department of Cardiovascular Medicine, Wenzhou Hospital of Traditional Chinese Medicine Wenzhou 325000, Zhejiang, China
| |
Collapse
|
13
|
Chen C, Lan L, Xu K. Remimazolam Combined with Andrographolide Improve Postoperative Cognitive Dysfunction in Rats after Cardiopulmonary Bypass through the AMPK/SIRT1 Signaling Pathway. J Integr Neurosci 2025; 24:25665. [PMID: 39862006 DOI: 10.31083/jin25665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 01/27/2025] Open
Abstract
INTRODUCTION The effects of remimazolam (Re) in combination with andrographolide (AP) on learning, memory, and motor abilities in rats following cardiopulmonary bypass (CPB) surgery were studied. METHODS We hypothesized that the combination of Re and AP could improve postoperative cognitive dysfunction (POCD) in rats after CPB by modulating nervous system inflammation. Cognitive function was assessed using the Morris Water Maze test, and the concentrations of tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6) in serum were measured by enzyme-linked immunosorbent assay (ELISA). Apoptosis was evaluated using western blotting and the terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) staining assay. RESULTS The results indicated that both Re and AP independently improved cognitive function in rats after CPB and inhibited the secretion of inflammatory factors and apoptosis in hippocampal tissues. Combined administration of Re and AP enhanced the alleviation of POCD compared with monotherapy. The adenosine monophosphate-activated protein kinase/silent information regulator of transcription 1 (AMPK/SIRT1) signaling pathway was activated by the combination of Re and AP. CONCLUSIONS Collectively, the combination of Re and AP treatment significantly improves POCD in rats after CPB through activation of the AMPK/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Chong Chen
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University, School of Medicine, 310009 Hangzhou, Zhejiang, China
| | - Lixia Lan
- Department of Anesthesia, Lishui Maternal and Child Health Center, 323000 Lishui, Zhejiang, China
| | - Kai Xu
- Department of Anesthesia, Hangzhou Plastic Surgery Hospital, 310000 Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Park M, Cho S, Jeong D. Restoration of Sestrin 3 Expression Mitigates Cardiac Oxidative Damage in Ischemia-Reperfusion Injury Model. Antioxidants (Basel) 2025; 14:61. [PMID: 39857395 PMCID: PMC11763094 DOI: 10.3390/antiox14010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Cardiac ischemia-reperfusion injury (IRI) occurs when blood flow is restored to the myocardium after a period of ischemia, leading to oxidative stress and subsequent myocardial cell damage, primarily due to the accumulation of reactive oxygen species (ROS). In our previous research, we identified that miR-25 is significantly overexpressed in pressure overload-induced heart failure, and its inhibition improves cardiac function by restoring the expression of SERCA2a, a key protein involved in calcium regulation. In this study, we aimed to investigate the role of miR-25 in the context of ischemia-reperfusion injury. We found that miR-25 was markedly upregulated under hypoxic conditions in both in vitro and in vivo models. Through in silico analysis, we identified Sestrin3 (SESN3), an antioxidant protein known for its protective effects against oxidative stress, as a novel target of miR-25. Based on these findings, we hypothesized that inhibiting miR-25 would restore Sestrin3 expression, thereby reducing ROS-induced myocardial cell damage and improving cardiac function. To test this hypothesis, we employed two model systems: a hypoxia/reoxygenation (H/R) stress model using H9c2 myoblasts and a surgically induced ischemia-reperfusion injury mouse model. Our results demonstrated that the use of miR-25 inhibitors significantly improved cardiac function and reduced myocardial damage in both models through the restoration of SESN3 expression. In conclusion, our findings suggest that targeting miR-25 may serve as a novel therapeutic modality to alleviate oxidative damage in the heart.
Collapse
Affiliation(s)
| | | | - Dongtak Jeong
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University—ERICA, Ansan 15588, Republic of Korea; (M.P.); (S.C.)
| |
Collapse
|
15
|
Yu X, Wang X, Xu F, Zhang X, Wang M, Zhou R, Sun Z, Pan X, Feng L, Zhang W, Sun Y, Zhang W, Zhou D, Jiang Y. Mir-615-3p promotes osteosarcoma progression via the SESN2/AMPK/mTOR pathway. Cancer Cell Int 2024; 24:411. [PMID: 39702297 DOI: 10.1186/s12935-024-03604-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common primary malignant bone neoplasm. Growing researches have highlighted the tumor promoting role of miR-615-3p in various cancers. Notwithstanding, the biological function and underlying mechanisms of miR-615-3p in OS development still unclear. METHODS Quantitative Real-Time PCR analysis (qRT-PCR) and RNA fluorescence in situ hybridization (FISH) staining were performed to measure miR-615-3p expression in OS. CCK-8 assay, colony formation assay and EdU assay were applied to analyze the OS cell proliferation activity. Cell metastasis abilities were evaluated using Transwell assays. Analysis of apoptosis was performed based on flow cytometric detection. The potential mechanisms of miR-615-3p in OS progression were investigated through RNA immunoprecipitation (RIP) assays, dual-luciferase reporter assays, qRT-PCR and western blotting. In vivo experiments, mouse xenograft model was carried out to assess the tumorigenicity of miR-615-3p. RESULTS This study demonstrated a significant upregulation of miR-615-3p in OS. In addition, miR-615-3p knockdown suppressed OS proliferation, invasion, metastasis and EMT. Mechanistically, miR-615-3p regulated sestrin 2 (SESN2) expression negatively by targeting its 3'UTR. Moreover, silencing SESN2 facilitated OS progression and activated mTOR pathway. Noteworthy, the anticancer functions of miR-615-3p knockdown were partially recovered by SESN2 silencing. Taken together, the miR-615-3p/SESN2/mTOR pathway is critical for regulating OS progression. CONCLUSION Our results revealed that miR-615-3p modulated mTOR signaling, thus influencing the progression of OS. For OS treatment, molecular strategies that target the miR-615-3p/SESN2/mTOR pathway is promising.
Collapse
Affiliation(s)
- Xuecheng Yu
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China
| | - Xin Wang
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China
| | - Fan Xu
- Department of Disease Control, 987 Hospital of Joint Logistics Support Force of PLA, Baoji, Shaanxi, China
| | - Xinyi Zhang
- Wenzhou Medical University, Wenzhou, 325035, China
| | - Muyi Wang
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China
| | - Ruikai Zhou
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China
| | - Zhengyi Sun
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China
| | - Xiaohui Pan
- Department of Orthopedics, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu Province, China
| | - Lin Feng
- The people's hospital of WuQia county, WuQia, 845450, Xinjiang, China
| | - Wanchao Zhang
- Department of Radiology, The people's hospital of WuQia county, WuQia, 845450, Xinjiang, China
| | - Yong Sun
- Department of Orthopedics, Wuqia People's Hospital, Xinjiang, China
| | - Wenting Zhang
- Affiliated Changzhou Children's Hospital of Nantong University, Changzhou, 213003, Jiangsu, China.
| | - Dong Zhou
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China.
- Department of Orthopedics, Wuqia People's Hospital, Xinjiang, China.
- Affiliated Changzhou Children's Hospital of Nantong University, Changzhou, 213003, Jiangsu, China.
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213003, China.
| | - Yuqing Jiang
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China.
| |
Collapse
|
16
|
Wang D, Jin Y, Yang M, Xue Y, Zhang X, Guo Y, Li X, Ma K. Cardioprotective effect of Saussurea involucrata injection against Doxorubicin-induced cardiotoxicity by network pharmacology analysis and experimental verification. Acta Biochim Biophys Sin (Shanghai) 2024; 57:554-568. [PMID: 39632659 PMCID: PMC12040744 DOI: 10.3724/abbs.2024170] [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: 05/06/2024] [Accepted: 07/10/2024] [Indexed: 12/07/2024] Open
Abstract
Doxorubicin (Dox) is widely utilized in the clinical treatment of various cancers. Despite its efficacy, Dox induces numerous adverse effects in humans with significant cardiotoxicity, posing a major limitation to its use. Saussurea involucrata injection (SII), derived from Saussurea involucrata, exhibits notable anti-inflammatory and anti-oxidative stress properties. However, its potential protective effects against Dox-induced cardiotoxicity (DIC) remain unexplored. In this study, we investigate the ability of SII to mitigate DIC and elucidate the underlying mechanisms through experimental research and network pharmacology analysis. Results from both in vitro and in vivo experiments reveal that SII treatment significantly improves Dox-induced cardiac dysfunction, reducing pathological alterations and fibrosis in cardiomyocytes. Moreover, SII has cardioprotective effects by diminishing the inflammation, oxidative stress, and apoptosis triggered by Dox. Network pharmacological analysis further shows that SII downregulates P53 protein expression by activating the AKT/MDM2 signaling pathway, thus attenuating DIC. In conclusion, this study confirms that SII mitigates DIC through downregulation of the AKT/MDM2/P53 signaling pathway, suggesting a promising therapeutic strategy for alleviating DIC.
Collapse
Affiliation(s)
- Ding Wang
- Key Laboratory of Xinjiang Endemic and Ethnic DiseasesMinistry of EducationShihezi University School of MedicineShihezi832003China
- Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated HospitalShihezi University School of MedicineShihezi832003China
- Department of PathophysiologyShihezi University School of MedicineShihezi832003China
| | - Yu Jin
- Key Laboratory of Xinjiang Endemic and Ethnic DiseasesMinistry of EducationShihezi University School of MedicineShihezi832003China
- Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated HospitalShihezi University School of MedicineShihezi832003China
- of PhysiologyShihezi University School of MedicineShihezi832003China
| | - Mengyu Yang
- Key Laboratory of Xinjiang Endemic and Ethnic DiseasesMinistry of EducationShihezi University School of MedicineShihezi832003China
- Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated HospitalShihezi University School of MedicineShihezi832003China
- of PhysiologyShihezi University School of MedicineShihezi832003China
| | - Yajing Xue
- Key Laboratory of Xinjiang Endemic and Ethnic DiseasesMinistry of EducationShihezi University School of MedicineShihezi832003China
- Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated HospitalShihezi University School of MedicineShihezi832003China
- of PhysiologyShihezi University School of MedicineShihezi832003China
| | - Xiaotong Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic DiseasesMinistry of EducationShihezi University School of MedicineShihezi832003China
- Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated HospitalShihezi University School of MedicineShihezi832003China
- of PhysiologyShihezi University School of MedicineShihezi832003China
| | - Yanli Guo
- Key Laboratory of Xinjiang Endemic and Ethnic DiseasesMinistry of EducationShihezi University School of MedicineShihezi832003China
- Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated HospitalShihezi University School of MedicineShihezi832003China
- of PhysiologyShihezi University School of MedicineShihezi832003China
| | - Xinzhi Li
- Key Laboratory of Xinjiang Endemic and Ethnic DiseasesMinistry of EducationShihezi University School of MedicineShihezi832003China
- Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated HospitalShihezi University School of MedicineShihezi832003China
- of PhysiologyShihezi University School of MedicineShihezi832003China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic DiseasesMinistry of EducationShihezi University School of MedicineShihezi832003China
- Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated HospitalShihezi University School of MedicineShihezi832003China
- of PhysiologyShihezi University School of MedicineShihezi832003China
| |
Collapse
|
17
|
Zhang S, Yang Y, Lv X, Zhou X, Zhao W, Meng L, Xu H, Zhu S, Wang Y. Doxorubicin-Induced Cardiotoxicity Through SIRT1 Loss Potentiates Overproduction of Exosomes in Cardiomyocytes. Int J Mol Sci 2024; 25:12376. [PMID: 39596439 PMCID: PMC11594621 DOI: 10.3390/ijms252212376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Mutual interaction between doxorubicin (DOX) and cardiomyocytes is crucial for cardiotoxicity progression. Cardiomyocyte injury is an important pathological feature of DOX-induced cardiomyopathy, and its molecular pathogenesis is multifaceted. In addition to the direct toxic effects of DOX on cardiomyocytes, DOX-induced exosomes in the extracellular microenvironment also regulate the pathophysiological states of cardiomyocytes. However, the mechanisms by which DOX regulates exosome secretion and subsequent pathogenesis remain incompletely understood. Here, we found that DOX significantly increased exosome secretion from cardiomyocytes, and inhibiting this release could alleviate cardiomyocyte injury. DOX promoted exosome secretion by reducing cardiomyocyte silencing information regulator 1 (SIRT1) expression, exacerbating cardiotoxicity. DOX impaired lysosomal acidification in cardiomyocytes, reducing the degradation of intracellular multivesicular bodies (MVBs), resulting in an increase in MVB volume before fusing with the plasma membrane to release their contents. Mechanistically, SIRT1 loss inhibited lysosomal acidification by reducing the expression of the ATP6V1A subunit of the lysosomal vacuolar-type H+ ATPase (V-ATPase) proton pump. Overexpressing SIRT1 increased ATP6V1A expression, improved lysosomal acidification, inhibited exosome secretion, and thereby alleviated DOX-induced cardiotoxicity. Interestingly, DOX also induced mitochondrial-derived vesicle formation in cardiomyocytes, which may further increase the abundance of MVBs and promote exosome release. Collectively, this study identified SIRT1-mediated impairment of lysosomal acidification as a key mechanism underlying the increased exosome secretion from cardiomyocytes induced by DOX, providing new insights into DOX-induced cardiotoxicity pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ying Wang
- Department of Forensic Medicine, School of Basic Medicine, Soochow University, Suzhou 215123, China; (S.Z.)
| |
Collapse
|
18
|
Safaie N, Idari G, Ghasemi D, Hajiabbasi M, Alivirdiloo V, Masoumi S, Zavvar M, Majidi Z, Faridvand Y. AMPK activation; a potential strategy to mitigate TKI-induced cardiovascular toxicity. Arch Physiol Biochem 2024:1-13. [PMID: 39526616 DOI: 10.1080/13813455.2024.2426494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/20/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The introduction of Tyrosine Kinase Inhibitors (TKIs) has revolutionised cancer treatment, yet concerns regarding cardiovascular toxicity have surfaced. This piece delves into the interplay between AMP-activated protein kinase (AMPK) signalling and TKI-induced cardiovascular toxicity. The study unravels the intricate relationship between AMPK activation and TKI-induced cardiovascular toxicity, aiming to ascertain whether AMPK can play a strategic role in mitigating adverse effects. Beyond unravelling mechanistic insights, the research sets the stage for future therapeutic approaches, envisioning AMPK activation as a pivotal connection for balancing effective cancer treatment with cardiovascular well-being. As research advances, the potential of AMPK activation not only addresses challenges in TKI-induced cardiovascular toxicity but also shapes the future landscape of personalised anticancer therapies. The article explores the mechanisms of TKI-induced toxicity, AMPK's impact on cardiovascular health, and the potential therapeutic implications of AMPK activation in alleviating TKI-associated toxicities.
Collapse
Affiliation(s)
- Nasser Safaie
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gholamreza Idari
- Department of Clinical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Diba Ghasemi
- Stem Cell research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Vahid Alivirdiloo
- Ramsar Campus, Mazandaran University of Medical Sciences, Ramasr, Iran
| | - Shahab Masoumi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Vanderbilt University of Medical center, Nashville, TN, USA
| | - Mahdi Zavvar
- Department of Medical Laboratory Science, School of Allied Medicine Sciences (SAMS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Ziba Majidi
- Department of Medical Laboratory Science, School of Allied Medicine Sciences (SAMS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Yousef Faridvand
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
19
|
Marín-Blázquez M, Rovira J, Ramírez-Bajo MJ, Zapata-Pérez R, Rabadán-Ros R. NAD + enhancers as therapeutic agents in the cardiorenal axis. Cell Commun Signal 2024; 22:537. [PMID: 39516787 PMCID: PMC11546376 DOI: 10.1186/s12964-024-01903-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Cardiorenal diseases represent a complex interplay between heart failure and renal dysfunction, being clinically classified as cardiorenal syndromes (CRS). Recently, the contributions of altered nicotinamide adenine dinucleotide (NAD+) metabolism, through deficient NAD+ synthesis and/or elevated consumption, have proved to be decisive in the onset and progress of cardiorenal disease. NAD+ is a pivotal coenzyme in cellular metabolism, being significant in various signaling pathways, such as energy metabolism, DNA damage repair, gene expression, and stress response. Convincing evidence suggests that strategies designed to boost cellular NAD+ levels are a promising therapeutic option to address cardiovascular and renal disorders. Here, we review and discuss the implications of NAD+ metabolism in cardiorenal diseases, focusing on the propitious NAD+ boosting therapeutic strategies, based on the use of NAD+ precursors, poly(ADP-ribose) polymerase inhibitors, sirtuin activators, and other alternative approaches, such as CD38 blockade, nicotinamide phosphoribosyltransferase activation and combined interventions.
Collapse
Affiliation(s)
- Mariano Marín-Blázquez
- Group of Metabolism and Genetic Regulation of Disease, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, 30107 Guadalupe de Maciascoque, Murcia, Spain
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Casanova 143 CRB CELLEX sector 2B, Barcelona, 08036, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), Madrid, Spain
| | - María José Ramírez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Casanova 143 CRB CELLEX sector 2B, Barcelona, 08036, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), Madrid, Spain
| | - Rubén Zapata-Pérez
- Group of Metabolism and Genetic Regulation of Disease, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, 30107 Guadalupe de Maciascoque, Murcia, Spain.
| | - Rubén Rabadán-Ros
- Group of Metabolism and Genetic Regulation of Disease, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, 30107 Guadalupe de Maciascoque, Murcia, Spain.
| |
Collapse
|
20
|
Law M, Wang PC, Zhou ZY, Wang Y. From Microcirculation to Aging-Related Diseases: A Focus on Endothelial SIRT1. Pharmaceuticals (Basel) 2024; 17:1495. [PMID: 39598406 PMCID: PMC11597311 DOI: 10.3390/ph17111495] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/23/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024] Open
Abstract
Silent information regulator sirtuin 1 (SIRT1) is an NAD+-dependent deacetylase with potent anti-arterial aging activities. Its protective function in aging-related diseases has been extensively studied. In the microcirculation, SIRT1 plays a crucial role in preventing microcirculatory endothelial senescence by suppressing inflammation and oxidative stress while promoting mitochondrial function and optimizing autophagy. It suppresses hypoxia-inducible factor-1α (HIF-1α)-mediated pathological angiogenesis while promoting healthy, physiological capillarization. As a result, SIRT1 protects against microvascular dysfunction, such as diabetic microangiopathy, while enhancing exercise-induced skeletal muscle capillarization and energy metabolism. In the brain, SIRT1 upregulates tight junction proteins and strengthens their interactions, thus maintaining the integrity of the blood-brain barrier. The present review summarizes recent findings on the regulation of microvascular function by SIRT1, the underlying mechanisms, and various approaches to modulate SIRT1 activity in microcirculation. The importance of SIRT1 as a molecular target in aging-related diseases, such as diabetic retinopathy and stroke, is underscored, along with the need for more clinical evidence to support SIRT1 modulation in the microcirculation.
Collapse
Affiliation(s)
- Martin Law
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.L.)
| | - Pei-Chun Wang
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.L.)
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Zhong-Yan Zhou
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.L.)
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yu Wang
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.L.)
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
21
|
Jiang M, Wang Y, Zhang J, Fan X, Jieensi M, Ding F, Wang Y, Sun X. Temperature and Ultrasound-Responsive Nanoassemblies for Enhanced Organ Targeting and Reduced Cardiac Toxicity. Int J Nanomedicine 2024; 19:11397-11413. [PMID: 39524922 PMCID: PMC11550713 DOI: 10.2147/ijn.s470465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Background Biocompatible nanocarriers are widely employed as drug-delivery vehicles for treatment. Nevertheless, indiscriminate drug release, insufficient organ-specific targeting, and systemic toxicity hamper nanocarrier effectiveness. Stimuli-responsive nano-sized drug delivery systems (DDS) are an important strategy for enhancing drug delivery efficiency and reducing unexpected drug release. Methods This study introduces a temperature- and ultrasound-responsive nano-DDS in which the copolymer p-(MEO2MA-co-THPMA) is grafted onto mesoporous iron oxide nanoparticles (MIONs) to construct an MPL-p nano-DDS. The copolymer acts as a nanopore gatekeeper, assuming an open conformation at sub-physiological temperatures that allows drug encapsulation and a closed conformation at physiological temperatures that prevents unexpected drug release during circulation. Lactoferrin was conjugated to the nanoparticle surface via polyethylene glycol to gain organ-targeting ability. External ultrasonic irradiation of the nanoparticles in the targeted organs caused a conformational change of the copolymer and reopened the pores, facilitating controlled drug release. Results MPL-p exhibited excellent biocompatibility and rare drug release in circulation. When targeting delivery to the brain, ultrasound promoted the release of the loaded drugs in the brain without accumulation in other organs, avoiding the related adverse reactions, specifically those affecting the heart. Conclusion This study established a novel temperature- and ultrasound-responsive DDS that reduced systemic adverse reactions compared with traditional DDS, especially in the heart, and demonstrated excellent organ delivery efficiency.
Collapse
Affiliation(s)
- Mingzhou Jiang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Yiming Wang
- Department of Cardiology, Huashan Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Jinjin Zhang
- Department of Cardiology, Huashan Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Xi Fan
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Milayi Jieensi
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Fang Ding
- Department of Cardiology, Huashan Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Yiqing Wang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Xiaotian Sun
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
22
|
Tang Z, Shu L, Cao Z, Xu Y, Li C. Osteoarthritis rat serum-derived extracellular vesicles aggravate osteoarthritis development by inducing NLRP3-mediated pyroptotic cell death and cellular inflammation. Hum Cell 2024; 37:1624-1637. [PMID: 39141224 DOI: 10.1007/s13577-024-01119-1] [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: 04/03/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
Osteoarthritis (OA), degenerative joint disease, is the most prevalent form of arthritis worldwide. Besides its substantial burden on society, the high OA morbidity greatly diminishes patients' quality of life. According to recent research, patients-derived serum extracellular vesicles (EVs) are critically involved in sustaining the corresponding disease progression. However, limited research has fully explored the specific functions and molecular mechanisms of OA serum-derived EVs in disease progression. Consequently, we aimed to investigate the underlying mechanism of OA rats-derived serum EVs in regulating OA progression. Before constructing the exosome-cell co-culture system, EVs were extracted from OA and control rat serum and co-cultured with bone marrow mesenchymal stem cells (BM-MSCs). Western blotting (WB), RT-qPCR, and enzyme-linked immunosorbent assay (ELISA) results revealed that OA rat serum-derived EVs upregulated cell pyroptosis-related markers, including nod-Like receptor protein-3 (NLRP3), apoptosis-associated speck-like protein (ASC), gasdermin D (GSDMD), and cleaved caspase-1. The OA rat-EVs also induced the release of LDH and inflammatory cytokines, including interleukin (IL)-1β, IL-18, IL-6, and TNF-α. Additional experiments revealed that OA rat-EVs delivered miR-133a-3p to BM-MSCs and upregulated miR-133a-3p to degrade sirtuin 1 (SIRT1), and activating the downstream NF-κB signaling pathway. Furthermore, the rescuing experiments confirmed that silencing SIRT1 abrogated the miR-133a-3p-induced protective effects in OA-EVs-treated BM-MSCs. In conclusion, OA rats-derived miR-133a-3p-containing EVs modulated the downstream SIRT1/NF-κB pathway-mediated pyroptotic cell death and inflammation in OA. In other words, this study confirmed the role and underlying mechanisms by which OA-associated serum EVs regulate pyroptosis and inflammation response in OA development.
Collapse
Affiliation(s)
- Zhifang Tang
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, No.212 Daguan Road, Xishan District, Kunming, 650032, Yunnan, China
| | - Longjun Shu
- The First People's Hospital of Dali City, Dali, 671000, China
| | - Zijian Cao
- Clinical Medical College of Dali University, Dali, 671000, China
| | - Yongqing Xu
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, No.212 Daguan Road, Xishan District, Kunming, 650032, Yunnan, China.
| | - Chuan Li
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, No.212 Daguan Road, Xishan District, Kunming, 650032, Yunnan, China.
| |
Collapse
|
23
|
Cui J, Chen Y, Yang Q, Zhao P, Yang M, Wang X, Mang G, Yan X, Wang D, Tong Z, Wang P, Kong Y, Wang N, Wang D, Dong N, Liu M, E M, Zhang M, Yu B. Protosappanin A Protects DOX-Induced Myocardial Injury and Cardiac Dysfunction by Targeting ACSL4/FTH1 Axis-Dependent Ferroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310227. [PMID: 38984448 PMCID: PMC11425893 DOI: 10.1002/advs.202310227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/12/2024] [Indexed: 07/11/2024]
Abstract
Doxorubicin (DOX) is an effective anticancer agent, but its clinical utility is constrained by dose-dependent cardiotoxicity, partly due to cardiomyocyte ferroptosis. However, the progress of developing cardioprotective medications to counteract ferroptosis has encountered obstacles. Protosappanin A (PrA), an anti-inflammatory compound derived from hematoxylin, shows potential against DOX-induced cardiomyopathy (DIC). Here, it is reported that PrA alleviates myocardial damage and dysfunction by reducing DOX-induced ferroptosis and maintaining mitochondrial homeostasis. Subsequently, the molecular target of PrA through proteome microarray, molecular docking, and dynamics simulation is identified. Mechanistically, PrA physically binds with ferroptosis-related proteins acyl-CoA synthetase long-chain family member 4 (ACSL4) and ferritin heavy chain 1 (FTH1), ultimately inhibiting ACSL4 phosphorylation and subsequent phospholipid peroxidation, while also preventing FTH1 autophagic degradation and subsequent release of ferrous ions (Fe2+) release. Given the critical role of ferroptosis in the pathogenesis of ischemia-reperfusion (IR) injury, this further investigation posits that PrA can confer a protective effect against IR-induced cardiac damage by inhibiting ferroptosis. Overall, a novel pharmacological inhibitor is unveiled that targets ferroptosis and uncover a dual-regulated mechanism for cardiomyocyte ferroptosis in DIC, highlighting additional therapeutic options for chemodrug-induced cardiotoxicity and ferroptosis-triggered disorders.
Collapse
|
24
|
Liu D, Cheng X, Wu H, Song H, Bu Y, Wang J, Zhang X, Yan C, Han Y. CREG1 attenuates doxorubicin-induced cardiotoxicity by inhibiting the ferroptosis of cardiomyocytes. Redox Biol 2024; 75:103293. [PMID: 39094399 PMCID: PMC11345695 DOI: 10.1016/j.redox.2024.103293] [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: 07/14/2024] [Accepted: 07/27/2024] [Indexed: 08/04/2024] Open
Abstract
OBJECTIVE Doxorubicin (DOX)-induced cardiotoxicity limits the application of DOX in cancer patients. Currently, there is no effective prevention or treatment for DOX-induced cardiotoxicity. The cellular repressor of E1A-stimulated genes (CREG1) is a cardioprotective factor that plays an important role in the maintenance of cardiomyocytes differentiation and homeostasis. However, the role and mechanism of CREG1 in DOX-induced cardiotoxicity has not yet been elucidated. METHODS In vivo, C57BL/6J mice, CREG1 transgenic and cardiac-specific CREG1 knockout mice were used to establish a DOX-induced cardiotoxicity model. H&E staining, Masson's trichrome, WGA staining, real-time PCR, and western blotting were performed to examine fibrosis and ferroptosis in the myocardium. In vitro, neonatal mouse cardiomyocytes (NMCMs) were cultured and stimulated with DOX, CREG1-overexpressed adenovirus, and small interfering RNA was used to establish CREG1 overexpression or knockdown cardiomyocytes. Transcriptomics, real-time PCR, western blotting, and immunoprecipitation were used to examine the roles and mechanisms of CREG1 in cardiomyocytes ferroptosis. RESULTS The mRNA and protein levels of CREG1 were reduced in the hearts and NMCMs after DOX treatment. CREG1 overexpression alleviated myocardial damage and inhibited DOX-induced ferroptosis in the myocardium. CREG1 deficiency in the heart aggravated DOX-induced cardiotoxicity and ferroptosis. In vitro, CREG1 overexpression inhibited cardiomyocytes ferroptosis induced by DOX, and CREG1 knockdown aggravated DOX-induced cardiotoxicity. Mechanistically, CREG1 inhibited the mRNA and protein expression of pyruvate dehydrogenase kinase 4 (PDK4) by regulating the F-box and WD repeat domain containing 7 (FBXW7)-forkhead box O1 (FOXO1) pathway. PDK4 deficiency reversed the effects of CREG1 knockdown on cardiomyocytes ferroptosis following DOX treatment. CONCLUSION CREG1 alleviated DOX-induced cardiotoxicity by inhibiting ferroptosis in cardiomyocytes. Our findings may help clarify the new roles of CREG1 in the development of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Dan Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaoli Cheng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China; Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hanlin Wu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Haixu Song
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yuxin Bu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jing Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaolin Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Yaling Han
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China.
| |
Collapse
|
25
|
Sun M, Zhang X, Tan B, Zhang Q, Zhao X, Dong D. Potential role of endoplasmic reticulum stress in doxorubicin-induced cardiotoxicity-an update. Front Pharmacol 2024; 15:1415108. [PMID: 39188945 PMCID: PMC11345228 DOI: 10.3389/fphar.2024.1415108] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/01/2024] [Indexed: 08/28/2024] Open
Abstract
As a chemotherapy agent, doxorubicin is used to combat cancer. However, cardiotoxicity has limited its use. The existing strategies fail to eliminate doxorubicin-induced cardiotoxicity, and an in-depth exploration of its pathogenesis is in urgent need to address the issue. Endoplasmic reticulum stress (ERS) occurs when Endoplasmic Reticulum (ER) dysfunction results in the accumulation of unfolded or misfolded proteins. Adaptive ERS helps regulate protein synthesis to maintain cellular homeostasis, while prolonged ERS stimulation may induce cell apoptosis, leading to dysfunction and damage to tissue and organs. Numerous studies on doxorubicin-induced cardiotoxicity strongly link excessive activation of the ERS to mechanisms including oxidative stress, calcium imbalance, autophagy, ubiquitination, and apoptosis. The researchers also found several clinical drugs, chemical compounds, phytochemicals, and miRNAs inhibited doxorubicin-induced cardiotoxicity by targeting ERS. The present review aims to outline the interactions between ERS and other mechanisms in doxorubicin-induced cardiotoxicity and summarize ERS's role in this type of cardiotoxicity. Additionally, the review enumerates several clinical drugs, phytochemicals, chemical compounds, and miRNAs targeting ERS for considering therapeutic regimens that address doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Xin Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Boxuan Tan
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China
| | - Qingya Zhang
- Innovation Institute, China Medical University, Shenyang, Liaoning, China
| | - Xiaopeng Zhao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Dan Dong
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
26
|
Lu L, Shao Y, Wang N, Xiong X, Zhai M, Tang J, Liu Y, Yang J, Yang L. Follistatin-like protein 1 attenuates doxorubicin-induced cardiomyopathy by inhibiting MsrB2-mediated mitophagy. Mol Cell Biochem 2024; 479:1817-1831. [PMID: 38696001 DOI: 10.1007/s11010-024-04955-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/30/2024] [Indexed: 07/18/2024]
Abstract
Doxorubicin (DOX) is a potent chemotherapeutic drug; however, its clinical use is limited due to its cardiotoxicity. Mitochondrial dysfunction plays a vital role in the pathogenesis of DOX-induced cardiomyopathy. Follistatin-like protein 1 (FSTL1) is a potent cardiokine that protects the heart from diverse cardiac diseases, such as myocardial infarction, cardiac ischemia/reperfusion injury, and heart failure. However, its role in DOX-induced cardiomyopathy is unclear. Therefore, the present study investigated whether administering recombinant FSTL1 could mitigate DOX-induced cardiomyopathy and clarified the underlying molecular mechanisms. FSTL1 treatment attenuated DOX-induced cardiac dysfunction, cardiac fibrosis, and cellular apoptosis by inhibiting excess mitochondrial matrix protein methionine sulfoxide reductase B2 (MsrB2)-mediated mitophagy. Furthermore, FSTL1 administration reduced the expression of apoptotic proteins, including MsrB2, Bax, caspase 3, mitochondrial Parkin, and LC3-II, increased myocardial ATP content, and decreased cardiac malondialdehyde levels, thus protecting mitochondrial function against DOX-induced cardiac injury. Furthermore, FSTL1 treatment protected the contractile properties of adult cardiomyocytes against DOX-induced injury in vitro. Furthermore, carbonyl cyanide m-chlorophenylhydrazone, a mitophagy inducer, impaired the protective effects of FSTL1 in DOX-treated H9c2 cardiomyocytes. In conclusion, these results show that FSTL1 is a novel therapeutic agent against DOX-induced cardiotoxicity that improves mitochondrial function and decreases mitophagy.
Collapse
Affiliation(s)
- Linhe Lu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yalan Shao
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Nisha Wang
- Department of Anesthesiology, Xi'an Children's Hospital, Xi'an Jiaotong University, Xi'an, 710003, China
| | - Xiang Xiong
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Mengen Zhai
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiayou Tang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yang Liu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jian Yang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lifang Yang
- Department of Anesthesiology, Xi'an Children's Hospital, Xi'an Jiaotong University, Xi'an, 710003, China.
| |
Collapse
|
27
|
Xie Y, Xie L, Qiu Z, He J, Jiang F, Cai M, Lin Y, Chen L. miR-485-3p targets SIRT1 in vascular smooth muscle cells mediating the occurrence of aortic dissection. J Cell Mol Med 2024; 28:e18454. [PMID: 39010253 PMCID: PMC11250145 DOI: 10.1111/jcmm.18454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 07/17/2024] Open
Abstract
Studies have demonstrated a close correlation between MicroRNA and the occurrence of aortic dissection (AD). However, the molecular mechanisms underlying this relationship have not been fully elucidated and further exploration is still required. In this study, we found that miR-485-3p was significantly upregulated in human aortic dissection tissues. Meanwhile, we constructed in vitro AD models in HAVSMCs, HAECs and HAFs and found that the expression of miR-485-3p was increased only in HAVSMCs. Overexpression or knockdown of miR-485-3p in HAVSMCs could regulate the expression of inflammatory cytokines IL1β, IL6, TNF-α, and NLRP3, as well as the expression of apoptosis-related proteins BAX/BCL2 and Cleaved caspase3/Caspase3. In the in vivo AD model, we have observed that miR-485-3p regulates vascular inflammation and apoptosis, thereby participating in the modulation of AD development in mice. Based on target gene prediction, we have validated that SIRT1 is a downstream target gene of miR-485-3p. Furthermore, by administering SIRT1 agonists and inhibitors to mice, we observed that the activation of SIRT1 alleviates vascular inflammation and apoptosis, subsequently reducing the incidence of AD. Additionally, functional reversal experiments revealed that overexpression of SIRT1 in HAVSMCs could reverse the cell inflammation and apoptosis mediated by miR-485-3p. Therefore, our research suggests that miR-485-3p can aggravate inflammation and apoptosis in vascular smooth muscle cells by suppressing the expression of SIRT1, thereby promoting the progression of aortic dissection.
Collapse
Affiliation(s)
- Yuling Xie
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouFujianP. R. China
- Key Laboratory of Cardio‐Thoracic Surgery (Fujian Medical University)Fujian Province UniversityFuzhouFujianP. R. China
| | - Linfeng Xie
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouFujianP. R. China
- Key Laboratory of Cardio‐Thoracic Surgery (Fujian Medical University)Fujian Province UniversityFuzhouFujianP. R. China
| | - Zhihuang Qiu
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouFujianP. R. China
- Key Laboratory of Cardio‐Thoracic Surgery (Fujian Medical University)Fujian Province UniversityFuzhouFujianP. R. China
| | - Jian He
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouFujianP. R. China
- Key Laboratory of Cardio‐Thoracic Surgery (Fujian Medical University)Fujian Province UniversityFuzhouFujianP. R. China
| | - Fei Jiang
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouFujianP. R. China
- Department of NursingFujian Medical University Union HospitalFuzhouFujianP. R. China
| | - Meiling Cai
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouFujianP. R. China
- Department of NursingFujian Medical University Union HospitalFuzhouFujianP. R. China
| | - Yanjuan Lin
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouFujianP. R. China
- Department of NursingFujian Medical University Union HospitalFuzhouFujianP. R. China
| | - Liangwan Chen
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouFujianP. R. China
- Key Laboratory of Cardio‐Thoracic Surgery (Fujian Medical University)Fujian Province UniversityFuzhouFujianP. R. China
| |
Collapse
|
28
|
Zhao X, Li M, Lu Y, Wang M, Xiao J, Xie Q, He X, Shuai S. Sirt1 inhibits macrophage polarization and inflammation in gouty arthritis by inhibiting the MAPK/NF-κB/AP-1 pathway and activating the Nrf2/HO-1 pathway. Inflamm Res 2024; 73:1173-1184. [PMID: 38739197 PMCID: PMC11214610 DOI: 10.1007/s00011-024-01890-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/13/2024] [Accepted: 05/04/2024] [Indexed: 05/14/2024] Open
Abstract
OBJECTIVE AND DESIGN To elucidate Sirt1's role in gouty arthritis inflammation and its potential mechanisms. MATERIAL Constructed murine models of gouty arthritis and conducted THP-1 cell experiments. TREATMENT 1 mg of MSU crystals injected into mice ankle joints for a 72-h intervention. After a 3-h pre-treatment with Sirt1-specific inhibitor (EX527) and agonist (SRT2104), inflammation was induced for 21 h using lipopolysaccharide (LPS) plus MSU crystals. METHODS We assessed gouty arthritis severity through joint inflammation index, swelling, and hematoxylin and eosin (H&E) staining, and measured CD68 mononuclear macrophages and Sirt1 expression in synovial tissue via immunohistochemistry. ELISA, NO assay, RT-qPCR, Flow cytometry, and Western blot were utilized to examine macrophage inflammatory factors, polarization, reactive oxygen species(ROS), MAPK/NF-κB/AP-1 and Nrf2/HO-1 pathways proteins. RESULTS Significant joint swelling, synovial tissue edema, and inflammatory cell infiltration were observed. CD68 mononuclear macrophages and Sirt1 expression were elevated in synovium. Sirt1 activation decreased inflammatory factors, M1 polarization, and ROS generation. Sirt1 activation reduced p38/JNK phosphorylation, thereby inhibiting downstream NF-κB p65/AP-1 and enhancing Nrf2/HO-1, thus suppressing inflammation. CONCLUSIONS Sirt1 alleviates M1 macrophage polarization and inflammation in gouty arthritis by inhibiting the MAPK/NF-κB/AP-1 pathway and activating the Nrf2/HO-1 pathway. Thus, activating Sirt1 may provide a new therapeutic target for gouty arthritis.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Menglan Li
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Yiwei Lu
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Mi Wang
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Jiawei Xiao
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Qingqing Xie
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xinyi He
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Shiquan Shuai
- Department of Rheumatology and Immunology, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, No. 97, Nanlu, Shunqing District, Nanchong, 637000, Sichuan, China.
- Nanchong Key Laboratory of Inflammation and Immunization, Nanchong Central Hospital (Nanchong Clinical Research Center), The Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
29
|
Bornaun T, Akkaya S, Güven HZ. Evaluation of Serum Sestrin 2 Levels in Patients Diagnosed with Endometrial Polyps and Uterine Leiomyomas. J Clin Med 2024; 13:3413. [PMID: 38929943 PMCID: PMC11204883 DOI: 10.3390/jcm13123413] [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: 04/14/2024] [Revised: 05/26/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Background/Objectives: This study investigates the correlation between the serum levels of Sestrin 2 and the presence of endometrial polyps or uterine leiomyomas, aiming to enhance the understanding of the pathophysiology underlying these gynecological conditions and evaluate the potential of Sestrin 2 as an early diagnostic biomarker. Methods: In a prospective case-control format, patients with preliminary diagnoses of endometrial polyps or uterine leiomyomas confirmed by histopathological analysis following surgery were included. This study analyzed serum Sestrin 2 levels across different patient groups, revealing significant variations that underscore the diagnostic value of Sestrin 2. Results: Elevated serum Sestrin 2 levels were observed in patients with endometrial polyps and uterine leiomyomas compared to the control group, suggesting its utility as a novel marker for early detection. Conclusions: The study indicates the promising role of serum Sestrin 2 levels as a valuable biomarker for early diagnosis of endometrial polyps and uterine leiomyomas, advocating for further research into its diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Teymur Bornaun
- Department of Obstetrics and Gynecology, Istanbul University Health Sciences Istanbul Bagcilar Training and Research Hospital, Istanbul 34200, Turkey
| | - Selim Akkaya
- Istanbul University Health Sciences Istanbul Bagcilar Training and Research Hospital, Istanbul 34200, Turkey; (S.A.); (H.Z.G.)
| | - Hamid Zafer Güven
- Istanbul University Health Sciences Istanbul Bagcilar Training and Research Hospital, Istanbul 34200, Turkey; (S.A.); (H.Z.G.)
| |
Collapse
|
30
|
Zhou M, Niu H, Cui D, Huang G, Li J, Tian H, Xu X, Liang F, Chen R. Resveratrol triggers autophagy-related apoptosis to inhibit the progression of colorectal cancer via inhibition of FOXQ1. Phytother Res 2024; 38:3218-3239. [PMID: 38682953 DOI: 10.1002/ptr.8184] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/14/2024] [Accepted: 02/27/2024] [Indexed: 05/01/2024]
Abstract
Colorectal cancer (CRC) is a significant health problem with elevated mortality rates, prompting intense exploration of its complex molecular mechanisms and innovative therapeutic avenues. Resveratrol (RSV), recognised for its anticancer effects through SIRT1 activation, is a promising candidate for CRC treatment. This study focuses on elucidating RSV's role in CRC progression, particularly its effect on autophagy-related apoptosis. Using bioinformatics, protein imprinting and immunohistochemistry, we established a direct correlation between FOXQ1 and adverse CRC prognosis. Comprehensive in vitro experiments confirmed RSV's ability to promote autophagy-related apoptosis in CRC cells. Plasmids for SIRT1 modulation were used to investigate underlying mechanisms. Molecular docking, glutathione-S-transferase pull-down experiments and immunoprecipitation highlighted RSV's direct activation of SIRT1, resulting in the inhibition of FOXQ1 expression. Downstream interventions identified ATG16L as a crucial autophagic target. In vivo and in vitro studies validated RSV's potential for CRC therapy through the SIRT1/FOXQ1/ATG16L pathway. This study establishes RSV's capacity to enhance autophagy-related cell apoptosis in CRC, positioning RSV as a prospective therapeutic agent for CRC within the SIRT1/FOXQ1/ATG16L pathway.
Collapse
Affiliation(s)
- MinFeng Zhou
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - HuiFang Niu
- School of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - DanDan Cui
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - GuoQuan Huang
- Department of Gastrointestinal Surgery, Enshi Central Hospital, Enshi City, China
| | - JinXiao Li
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - HaoRan Tian
- School of Acupuncture and Bone Injury, Hubei University of Chinese Medicine, Wuhan, China
| | - XiaoJuan Xu
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - FengXia Liang
- School of Acupuncture and Bone Injury, Hubei University of Chinese Medicine, Wuhan, China
| | - Rui Chen
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
李 然, 王 振, 王 燕, 滕 菲. [Sanshentongmai Mixture Improves Oxidative Damage in Rat Cardiomyocytes H9C2 via Upregulation of microRNA-146a]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:630-634. [PMID: 38948270 PMCID: PMC11211798 DOI: 10.12182/20240560601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Indexed: 07/02/2024]
Abstract
Objective To investigate the effect of Sanshentongmai (SSTM) mixture on the regulation of oxidative damage to rat cardiomyocytes (H9C2) through microRNA-146a and its mechanism. Methods H9C2 were cultured in vitro, H2O2 was used as an oxidant to create an oxidative damage model in H9C2 cells. SSTM intervention was administered to the H9C2 cells. Then, the changes in H2O2-induced oxidative damage in H9C2 cells and the expression of microRNA-146a were observed to explore the protective effect of SSTM on H9C2 and its mechanism. H9C2 cells cultured i n vitro were divided into 3 groups, including a control group, a model group of H2O2-induced oxidative damage (referred to hereafter as the model group), and a group given H2O2 modeling plus SSTM intervention at 500 μg/L for 72 h (referred to hereafter as the treatment group). The cell viability was measured by CCK8 assay. In addition, the levels of N-terminal pro-brain natriuretic peptide (Nt-proBNP), nitric oxide (NO), high-sensitivity C-reactive protein (Hs-CRP), and angiotensin were determined by enzyme-linked immunosorbent assay (ELISA). The expression level of microRNA-146a was determined by real-time PCR (RT-PCR). Result H9C2 cells were pretreated with SSTM at mass concentrations ranging from 200 to 1500 μg/L. Then, CCK8 assay was performed to measure cell viability and the findings showed that the improvement in cell proliferation reached its peak when the mass concentration of SSTM was 500 μg/L, which was subsequently used as the intervention concentration. ELISA was performed to measure the indicators related to heart failure, including Nt-proBNP, NO, Hs-CRP, and angiotensin Ⅱ. Compared with those of the control group, the expressions of Nt-proBNP and angiotensin Ⅱ in the treatment group were up-regulated (P<0.05), while the expression of NO was down-regulated (P<0.05). There was no significant difference in the expression of Hs-CRP between the treatment group and the control group. These findings indicate that SSTM could effectively ameliorate oxidative damage in H9C2 rat cardiomyocytes. Finally, according to the RT-PCR findings for the expression of microRNA-146a in each group, H2O2 treatment at 15 μmol/L could significantly reduce the expression of microRNA-146a, and the expression of microRNA-146a in the treatment group was nearly doubled compared with that in the model group. There was no significant difference between the treatment group and the control group. Conclusion SSTM can significantly resist the H2O2-induced oxidative damage of H9C2 cells and may play a myocardial protective role by upregulating microRNA-146a.
Collapse
Affiliation(s)
- 然 李
- 首都医科大学附属北京中医医院 干部保健科 (北京 100010)Department of Health Care for Cadres, Beijing Hospital of Traditional Chinese Medicine Affiliated to the Capital Medical University, Beijing 100010, China
| | - 振裕 王
- 首都医科大学附属北京中医医院 干部保健科 (北京 100010)Department of Health Care for Cadres, Beijing Hospital of Traditional Chinese Medicine Affiliated to the Capital Medical University, Beijing 100010, China
| | - 燕丽 王
- 首都医科大学附属北京中医医院 干部保健科 (北京 100010)Department of Health Care for Cadres, Beijing Hospital of Traditional Chinese Medicine Affiliated to the Capital Medical University, Beijing 100010, China
| | - 菲 滕
- 首都医科大学附属北京中医医院 干部保健科 (北京 100010)Department of Health Care for Cadres, Beijing Hospital of Traditional Chinese Medicine Affiliated to the Capital Medical University, Beijing 100010, China
| |
Collapse
|
32
|
Zhang S, Zheng B, Wei Y, Liu Y, Yang L, Qiu Y, Su J, Qiu M. Bioinspired ginsenoside Rg3 PLGA nanoparticles coated with tumor-derived microvesicles to improve chemotherapy efficacy and alleviate toxicity. Biomater Sci 2024; 12:2672-2688. [PMID: 38596867 DOI: 10.1039/d4bm00159a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Breast cancer, a pervasive malignancy affecting women, demands a diverse treatment approach including chemotherapy, radiotherapy, and surgical interventions. However, the effectiveness of doxorubicin (DOX), a cornerstone in breast cancer therapy, is limited when used as a monotherapy, and concerns about cardiotoxicity persist. Ginsenoside Rg3, a classic compound of traditional Chinese medicine found in Panax ginseng C. A. Mey., possesses diverse pharmacological properties, including cardiovascular protection, immune modulation, and anticancer effects. Ginsenoside Rg3 is considered a promising candidate for enhancing cancer treatment when combined with chemotherapy agents. Nevertheless, the intrinsic challenges of Rg3, such as its poor water solubility and low oral bioavailability, necessitate innovative solutions. Herein, we developed Rg3-PLGA@TMVs by encapsulating Rg3 within PLGA nanoparticles (Rg3-PLGA) and coating them with membranes derived from tumor cell-derived microvesicles (TMVs). Rg3-PLGA@TMVs displayed an array of favorable advantages, including controlled release, prolonged storage stability, high drug loading efficiency and a remarkable ability to activate dendritic cells in vitro. This activation is evident through the augmentation of CD86+CD80+ dendritic cells, along with a reduction in phagocytic activity and acid phosphatase levels. When combined with DOX, the synergistic effect of Rg3-PLGA@TMVs significantly inhibits 4T1 tumor growth and fosters the development of antitumor immunity in tumor-bearing mice. Most notably, this delivery system effectively mitigates the toxic side effects of DOX, particularly those affecting the heart. Overall, Rg3-PLGA@TMVs provide a novel strategy to enhance the efficacy of DOX while simultaneously mitigating its associated toxicities and demonstrate promising potential for the combined chemo-immunotherapy of breast cancer.
Collapse
Affiliation(s)
- Shulei Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Bo Zheng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yiqi Wei
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yuhao Liu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Lan Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yujiao Qiu
- The Wharton School and School of Nursing, University of Pennsylvania, 19104, Philadelphia, USA
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
33
|
Wen F, Xu A, Wei W, Yang S, Xi Z, Ge Y, Wu S, Ju Z. Nicotinamide Mononucleotide Supplementation Alleviates Doxorubicin-Induced Multi-Organ Fibrosis. Int J Mol Sci 2024; 25:5303. [PMID: 38791345 PMCID: PMC11120852 DOI: 10.3390/ijms25105303] [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: 04/15/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Doxorubicin (DOX) is a potent chemotherapeutic agent known for its multi-organ toxicity, especially in the heart, which limits its clinical application. The toxic side effects of DOX, including DNA damage, oxidative stress, mitochondrial dysfunction and cell apoptosis, are intricately linked to the involvement of nicotinamide adenine dinucleotide (NAD+). To assess the effectiveness of the NAD+ precursor nicotinamide mononucleotide (NMN) in counteracting the multi-organ toxicity of DOX, a mouse model was established through DOX administration, which led to significant reductions in NAD+ in tissues with evident injury, including the heart, liver and lungs. NMN treatment alleviated both multi-organ fibrosis and mortality in mice. Mechanistically, tissue fibrosis, macrophage infiltration and DOX-related cellular damage, which are potentially implicated in the development of multi-organ fibrosis, could be attenuated by NAD+ restoration. Our findings provide compelling evidence for the benefits of NMN supplementation in mitigating the adverse effects of chemotherapeutic drugs on multiple organs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shu Wu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (F.W.); (Y.G.)
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (F.W.); (Y.G.)
| |
Collapse
|
34
|
Gao T, Wang J, Xiao M, Wang J, Wang S, Tang Y, Zhang J, Lu G, Guo H, Guo Y, Liu Q, Li J, Gu J. SESN2-Mediated AKT/GSK-3β/NRF2 Activation to Ameliorate Adriamycin Cardiotoxicity in High-Fat Diet-Induced Obese Mice. Antioxid Redox Signal 2024; 40:598-615. [PMID: 37265150 DOI: 10.1089/ars.2022.0156] [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] [Indexed: 06/03/2023]
Abstract
Aims: Obese patients are highly sensitive to adriamycin (ADR)-induced cardiotoxicity. However, the potential mechanism of superimposed toxicity remains to be elucidated. Sestrin 2 (SESN2), a potential antioxidant, could attenuate stress-induced cardiomyopathy; therefore, this study aims to explore whether SESN2 enhances cardiac resistance to ADR-induced oxidative damage in high-fat diet (HFD)-induced obese mice. Results: The results revealed that obesity decreased SESN2 expression in ADR-exposed heart. And, HFD mice may predispose to ADR-induced cardiotoxicity, which was probably associated with inhibiting protein kinase B (AKT), glycogen synthase kinase-3 beta (GSK-3β) phosphorylation and subsequently blocking nuclear localization of nuclear factor erythroid-2 related factor 2 (NRF2), ultimately resulting in cardiac oxidative damage. However, these destructive cascades and cardiac oxidative damage effects induced by HFD/sodium palmitate combined with ADR were blocked by overexpression of SESN2. Moreover, the antioxidant effect of SESN2 could be largely abolished by sh-Nrf2 or wortmannin. And sulforaphane, an NRF2 agonist, could remarkably reverse cardiac pathological and functional abnormalities caused by ADR in obese mice. Innovation and Conclusion: This study demonstrated that SESN2 might be a promising therapeutic target for improving anthracycline-related cardiotoxicity in obesity by upregulating activity of NRF2 via AKT/GSK-3β/Src family tyrosine kinase signaling pathway. Antioxid. Redox Signal. 40, 598-615.
Collapse
Affiliation(s)
- Ting Gao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mengjie Xiao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shudong Wang
- Department of Cardiology at the First Hospital of Jilin University, Changchun, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Jingjing Zhang
- Department of Cardiology at the First Hospital of China Medical University, Shenyang, China
- Department of Cardiology at the People's Hospital of Liaoning Province, Shenyang, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hua Guo
- Department of Nursing, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qingbo Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiahao Li
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
35
|
Zeng H, Zou P, Chen Y, Zhang P, Shao L. NOX4 aggravates doxorubicin-induced cardiomyocyte pyroptosis by increasing reactive oxygen species content and activating the NLRP3 inflammasome. Cardiovasc Diagn Ther 2024; 14:84-100. [PMID: 38434559 PMCID: PMC10904297 DOI: 10.21037/cdt-23-142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 11/24/2023] [Indexed: 03/05/2024]
Abstract
Background Nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4)-mediated reactive oxygen species (ROS) has been reported to induce cardiomyocyte apoptosis, but its effect on pyroptosis of cardiomyocytes has been rarely reported. This paper aimed to explore the effects of NOX4-mediated ROS production on doxorubicin (DOX)-induced myocardial injury and pyroptosis through nucleotide-binding and oligomerization domain-like receptor protein 3 (NLRP3) inflammasome. Methods HL-1 cells were treated with DOX or mice (30 mice were divided into five groups with six mice/group) underwent intraperitoneal injection with DOX (5 mg/kg, once a week, five times) to induce myocardial injury, followed by assessment of NOX4 and NLRP3 expression in cell supernatant and myocardial tissues. In cardiomyocyte HL-1 cells, cell proliferation was tested by MTT assay and the activity of ROS by probes. The superoxide dismutase (SOD) activity, malondialdehyde (MDA) content, and glutathione (GSH) activity were evaluated by kits. The expression of pyroptosis proteins was assessed by western blotting. Subsequently, the expression of NOX4 or NLRP3 was altered to determine the effect of NOX4 or NLRP3 expression on cardiomyocyte injury and pyroptosis. The animal models were utilized to evaluate the changes in the cardiac function of mice using an echocardiographic system, with these parameters measured including left ventricular ejection fraction (LVEF), left ventricular fractional shortening (LVFS), and left ventricular end-diastolic diameter (LVEDD). Furthermore, the content of myocardial injury markers and the protein expression of pyroptosis proteins were determined to evaluate myocardial injury in the mice. Results DOX treatment led to cardiomyocyte injury and pyroptosis, as evidenced by weakened LVEF, LVFS, and cell proliferation (P<0.05), elevated LVEDD, ROS, and MDA (P<0.05), increased expression of pyroptosis proteins (P<0.05), and decreased SOD and GSH (P<0.05). Additionally, NOX4 and NLRP3 were highly-expressed (P<0.05) in cell supernatant and myocardial tissues. In DOX-induced HL-1 cells, the overexpression of NOX4 intensified ROS levels to aggravate cardiomyocyte injury and pyroptosis, which was reversed by treatment of the ROS scavenger N-acetyl-cysteine. Furthermore, it was revealed that the combination of short hairpin RNA (sh)-NOX4 and overexpressed (oe)-NLRP3 reversed the cardioprotective effects of sh-NOX4 and increased myocardial tissue or cell injury and pyroptosis in vitro and in vivo. No mice died during the animal experiments, and only two were ruled out due to a weight loss greater than 20%. Conclusions NOX4-mediated ROS production activated NLRP3 inflammasome, thereby aggravating DOX-induced myocardial injury in vitro and in vivo.
Collapse
Affiliation(s)
- Hong Zeng
- Department of Cardiology, Jiangxi Provincial People’s Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Pengtao Zou
- Department of Cardiology, Jiangxi Provincial People’s Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yanmei Chen
- Department of Cardiology, Jiangxi Provincial People’s Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Ping Zhang
- Department of Neurology, Jiangxi Provincial People’s Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Liang Shao
- Department of Cardiology, Jiangxi Provincial People’s Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
36
|
Luan Y, Zhu X, Jiao Y, Liu H, Huang Z, Pei J, Xu Y, Yang Y, Ren K. Cardiac cell senescence: molecular mechanisms, key proteins and therapeutic targets. Cell Death Discov 2024; 10:78. [PMID: 38355681 PMCID: PMC10866973 DOI: 10.1038/s41420-023-01792-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Cardiac aging, particularly cardiac cell senescence, is a natural process that occurs as we age. Heart function gradually declines in old age, leading to continuous heart failure, even in people without a prior history of heart disease. To address this issue and improve cardiac cell function, it is crucial to investigate the molecular mechanisms underlying cardiac senescence. This review summarizes the main mechanisms and key proteins involved in cardiac cell senescence. This review further discusses the molecular modulators of cellular senescence in aging hearts. Furthermore, the discussion will encompass comprehensive descriptions of the key drugs, modes of action and potential targets for intervention in cardiac senescence. By offering a fresh perspective and comprehensive insights into the molecular mechanisms of cardiac senescence, this review seeks to provide a fresh perspective and important theoretical foundations for the development of drugs targeting this condition.
Collapse
Affiliation(s)
- Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Xiaofan Zhu
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Yuxue Jiao
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Hui Liu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, P. R. China
| | - Zhen Huang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, P. R. China
| | - Jinyan Pei
- Quality Management Department, Henan No.3 Provincial People's Hospital, Zhengzhou, 450052, P. R. China
| | - Yawei Xu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052, P. R. China.
| |
Collapse
|
37
|
Zhou N, Wei S, Sun T, Xie S, Liu J, Li W, Zhang B. Recent progress in the role of endogenous metal ions in doxorubicin-induced cardiotoxicity. Front Pharmacol 2023; 14:1292088. [PMID: 38143497 PMCID: PMC10748411 DOI: 10.3389/fphar.2023.1292088] [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: 09/11/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023] Open
Abstract
Doxorubicin is a widely used anticancer drug in clinical practice for the treatment of various human tumors. However, its administration is associated with cardiotoxicity. Administration of doxorubicin with low side effects for cancer treatment and prevention are, accordingly, urgently required. The human body harbors various endogenous metal ions that exert substantial influences. Consequently, extensive research has been conducted over several decades to investigate the potential of targeting endogenous metal ions to mitigate doxorubicin's side effects and impede tumor progression. In recent years, there has been a growing body of research indicating the potential efficacy of metal ion-associated therapeutic strategies in inhibiting doxorubicin-induced cardiotoxicity (DIC). These strategies offer a combination of favorable safety profiles and potential clinical utility. Alterations in intracellular levels of metal ions have been found to either facilitate or mitigate the development of DIC. For instance, ferroptosis, a cellular death mechanism, and metal ions such as copper, zinc, and calcium have been identified as significant contributors to DIC. This understanding can contribute to advancements in cancer treatment and provide valuable insights for mitigating the cardiotoxic effects of other therapeutic drugs. Furthermore, potential therapeutic strategies have been investigated to alleviate DIC in clinical settings. The ultimate goal is to improve the efficacy and safety of Dox and offer valuable insights for future research in this field.
Collapse
Affiliation(s)
- Ni Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
- School of Pharmacy, Central South University, Changsha, Hunan, China
| | - Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Taoli Sun
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Suifen Xie
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
- School of Pharmacy, Central South University, Changsha, Hunan, China
| | - Jian Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
- School of Pharmacy, Central South University, Changsha, Hunan, China
| |
Collapse
|
38
|
Chaulin AM. The Essential Strategies to Mitigate Cardiotoxicity Caused by Doxorubicin. Life (Basel) 2023; 13:2148. [PMID: 38004288 PMCID: PMC10672543 DOI: 10.3390/life13112148] [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: 01/12/2023] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 11/26/2023] Open
Abstract
The study of mechanisms underlying cardiotoxicity of doxorubicin and the development of strategies to mitigate doxorubicin-induced cardiotoxicity are the most relevant issues of modern cardio-oncology. This is due to the high prevalence of cancer in the population and the need for frequent use of highly effective chemotherapeutic agents, in particular anthracyclines, for optimal management of cancer patients. However, while being a potent agent to counteract cancer, doxorubicin also affects the cardiovascular systems of patients undergoing chemotherapy in a significant and unfavorable fashion. Consecutively reviewed in this article are risk factors and mechanisms of doxorubicin cardiotoxicity, and the essential strategies to mitigate cardiotoxic effects of doxorubicin treatment in cancer patients are discussed.
Collapse
Affiliation(s)
- Aleksey Michailovich Chaulin
- Department of Histology and Embryology, Samara State Medical University, Samara 443099, Russia;
- Department of Clinical Chemistry, Samara State Medical University, Samara 443099, Russia
| |
Collapse
|
39
|
Feng S, Tang D, Wang Y, Li X, Bao H, Tang C, Dong X, Li X, Yang Q, Yan Y, Yin Z, Shang T, Zheng K, Huang X, Wei Z, Wang K, Qi S. The mechanism of ferroptosis and its related diseases. MOLECULAR BIOMEDICINE 2023; 4:33. [PMID: 37840106 PMCID: PMC10577123 DOI: 10.1186/s43556-023-00142-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023] Open
Abstract
Ferroptosis, a regulated form of cellular death characterized by the iron-mediated accumulation of lipid peroxides, provides a novel avenue for delving into the intersection of cellular metabolism, oxidative stress, and disease pathology. We have witnessed a mounting fascination with ferroptosis, attributed to its pivotal roles across diverse physiological and pathological conditions including developmental processes, metabolic dynamics, oncogenic pathways, neurodegenerative cascades, and traumatic tissue injuries. By unraveling the intricate underpinnings of the molecular machinery, pivotal contributors, intricate signaling conduits, and regulatory networks governing ferroptosis, researchers aim to bridge the gap between the intricacies of this unique mode of cellular death and its multifaceted implications for health and disease. In light of the rapidly advancing landscape of ferroptosis research, we present a comprehensive review aiming at the extensive implications of ferroptosis in the origins and progress of human diseases. This review concludes with a careful analysis of potential treatment approaches carefully designed to either inhibit or promote ferroptosis. Additionally, we have succinctly summarized the potential therapeutic targets and compounds that hold promise in targeting ferroptosis within various diseases. This pivotal facet underscores the burgeoning possibilities for manipulating ferroptosis as a therapeutic strategy. In summary, this review enriched the insights of both investigators and practitioners, while fostering an elevated comprehension of ferroptosis and its latent translational utilities. By revealing the basic processes and investigating treatment possibilities, this review provides a crucial resource for scientists and medical practitioners, aiding in a deep understanding of ferroptosis and its effects in various disease situations.
Collapse
Affiliation(s)
- Shijian Feng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Dan Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yichang Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiang Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Bao
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengbing Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiuju Dong
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xinna Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qinxue Yang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yun Yan
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhijie Yin
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tiantian Shang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Kaixuan Zheng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaofang Huang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zuheng Wei
- Chengdu Jinjiang Jiaxiang Foreign Languages High School, Chengdu, People's Republic of China
| | - Kunjie Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Shiqian Qi
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
40
|
Gu X, Zhao L, Ye J, Chen L, Sui C, Li B, Wang X, Zhang J, Du Y. 1,25(OH) 2D 3 ameliorates doxorubicin‑induced cardiomyopathy by inhibiting the NLRP3 inflammasome and oxidative stress. Exp Ther Med 2023; 26:413. [PMID: 37559932 PMCID: PMC10407981 DOI: 10.3892/etm.2023.12112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/16/2023] [Indexed: 08/11/2023] Open
Abstract
Doxorubicin (DOX), as a chemotherapy agent with marked therapeutic effect, can be used to treat certain types of cancer such as leukemia, lymphoma and breast cancer. However, the toxic effects of DOX on cardiomyocytes limit its clinical application. Oxidative stress has been documented to serve a pivotal role in DOX-induced cardiomyopathy. Previous studies have reported that 1,25(OH)2D3 has antioxidant and anti-inflammatory effects and can inhibit the renin-angiotensin system. However, the effects of 1,25(OH)2D3 on the pathophysiological processes of DOX-induced cardiomyopathy and its mechanisms remain poorly understood. To investigate these potential effects, C57BL/6J mice were used to construct a DOX-induced cardiomyopathy model and treated with 1,25(OH)2D3. At 4 weeks after the first injection of DOX, cardiac function and myocardial injury were evaluated by echocardiograph and ELISA. Masson's trichrome staining and RT-qPCR were used to assess myocardial fibrosis, and immunohistochemistry and western blotting were performed to analyze expression levels of inflammation and oxidative stress, and the NLRP3 inflammasome pathway. ChIP assay was used to assess the effects of 1,25(OH)2D3 on histone modification in the NLRP3 and Nrf2 promoters. The results showed that 1,25(OH)2D3 treatment increased LVEF and LVFS, reduced serum levels of BNP and cTnT, inhibited the collagen deposition and profibrotic molecular expression, and downregulated the levels of inflammatory cytokines in DOX-induced cardiomyopathy. ROS and antioxidant indices were also ameliorated after 1,25(OH)2D3 treatment. In addition, 1,25(OH)2D3 was found to inhibit the NLRP3 inflammasome and KEAP-Nrf2 pathways through regulation of the levels of H3K4me3, H3K27me3 and H2AK119Ub in the NLRP3 and Nrf2 promoters. In conclusion, the present study demonstrated that 1,25(OH)2D3 regulated histone modification in the NLRP3 and Nrf2 promoters, which in turn inhibits the activation of NLRP3 inflammasome and oxidative stress in cardiomyocytes, alleviating DOX-induced cardiomyopathy. Therefore, 1,25(OH)2D3 may be a potential drug candidate for the treatment of DOX-induced cardiomyopathy.
Collapse
Affiliation(s)
- Xin Gu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Lin Zhao
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Jiabao Ye
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, P.R. China
| | - Lin Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, P.R. China
| | - Chenyan Sui
- Department of Neurology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Baihong Li
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Xiaoyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Jun Zhang
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, P.R. China
| | - Yingqiang Du
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, P.R. China
| |
Collapse
|
41
|
Basak M, Das K, Mahata T, Kumar D, Nagar N, Poluri KM, Kumar P, Das P, Stewart A, Maity B. RGS7 balances acetylation/de-acetylation of p65 to control chemotherapy-dependent cardiac inflammation. Cell Mol Life Sci 2023; 80:255. [PMID: 37589751 PMCID: PMC11071981 DOI: 10.1007/s00018-023-04895-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/28/2023] [Accepted: 07/22/2023] [Indexed: 08/18/2023]
Abstract
Cardiotoxicity remains a major limitation in the clinical utility of anthracycline chemotherapeutics. Regulator of G-protein Signaling 7 (RGS7) and inflammatory markers are up-regulated in the hearts of patients with a history of chemotherapy particularly those with reduced left-ventricular function. RGS7 knockdown in either the murine myocardium or isolated murine ventricular cardiac myocytes (VCM) or cultured human VCM provided marked protection against doxorubicin-dependent oxidative stress, NF-κB activation, inflammatory cytokine production, and cell death. In exploring possible mechanisms causally linking RGS7 to pro-inflammatory signaling cascades, we found that RGS7 forms a complex with acetylase Tip60 and deacetylase sirtuin 1 (SIRT1) and controls the acetylation status of the p65 subunit of NF-κB. In VCM, the detrimental impact of RGS7 could be mitigated by inhibiting Tip60 or activating SIRT1, indicating that the ability of RGS7 to modulate cellular acetylation capacity is critical for its pro-inflammatory actions. Further, RGS7-driven, Tip60/SIRT1-dependent cytokines released from ventricular cardiac myocytes and transplanted onto cardiac fibroblasts increased oxidative stress, markers of transdifferentiation, and activity of extracellular matrix remodelers emphasizing the importance of the RGS7-Tip60-SIRT1 complex in paracrine signaling in the myocardium. Importantly, while RGS7 overexpression in heart resulted in sterile inflammation, fibrotic remodeling, and compromised left-ventricular function, activation of SIRT1 counteracted the detrimental impact of RGS7 in heart confirming that RGS7 increases acetylation of SIRT1 substrates and thereby drives cardiac dysfunction. Together, our data identify RGS7 as an amplifier of inflammatory signaling in heart and possible therapeutic target in chemotherapeutic drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Madhuri Basak
- Centre of Biomedical Research (CBMR), SGPGI, SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Kiran Das
- Centre of Biomedical Research (CBMR), SGPGI, SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Tarun Mahata
- Centre of Biomedical Research (CBMR), SGPGI, SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Dinesh Kumar
- Centre of Biomedical Research (CBMR), SGPGI, SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Nupur Nagar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Pranesh Kumar
- Institute of Pharmaceutical Sciences, University of Lucknow, Lucknow, Uttar Pradesh, 226025, India
| | - Priyadip Das
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamilnadu, 603203, India
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA.
| | - Biswanath Maity
- Centre of Biomedical Research (CBMR), SGPGI, SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India.
| |
Collapse
|
42
|
Zhuang Y, Xu J, Zheng K, Zhang H. Research progress of postoperative cognitive dysfunction in cardiac surgery under cardiopulmonary bypass. IBRAIN 2023; 10:290-304. [PMID: 39346790 PMCID: PMC11427806 DOI: 10.1002/ibra.12123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 10/01/2024]
Abstract
Cardiopulmonary bypass (CPB) is often used in cardiothoracic surgery because its nonphysiological state causes pathophysiological changes in the body, causing multiorgan and multitissue damage to varying degrees. Postoperative cognitive dysfunction (POCD) is a common central nervous system complication after cardiac surgery. The etiology and mechanism of POCD are not clear. Neuroinflammation, brain mitochondrial dysfunction, cerebral embolism, ischemia, hypoxia, and other factors are related to the pathogenesis of POCD. There is a close relationship between CPB and POCD, as CPB can cause inflammation, hypoxia and reperfusion injury, and microemboli formation, all of which can trigger POCD. POCD increases medical costs, seriously affects patients' quality of life, and increases mortality. Currently, there is a lack of effective treatment methods for POCD. Commonly used methods include preoperative health management, reducing inflammation response during surgery, preventing microemboli formation, and implementing individualized rehabilitation programs after surgery. Strengthening preventive measures can minimize the occurrence of POCD and its adverse effects.
Collapse
Affiliation(s)
- Yi‐Ming Zhuang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Ji‐Yang Xu
- Department of AnesthesiologyJudicial Police Hospital of Guizhou ProvinceGuiyangChina
| | - Kun Zheng
- Department of AnesthesiologyGuizhou Provincial People's HospitalGuiyangChina
| | - Hong Zhang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| |
Collapse
|
43
|
Lee EJ, Jang WB, Choi J, Lim HJ, Park S, Rethineswaran VK, Ha JS, Yun J, Hong YJ, Choi YJ, Kwon SM. The Protective Role of Glutathione against Doxorubicin-Induced Cardiotoxicity in Human Cardiac Progenitor Cells. Int J Mol Sci 2023; 24:12070. [PMID: 37569446 PMCID: PMC10419046 DOI: 10.3390/ijms241512070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/22/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
This study investigated the protective effect of glutathione (GSH), an antioxidant drug, against doxorubicin (DOX)-induced cardiotoxicity. Human cardiac progenitor cells (hCPCs) treated with DOX (250 to 500 nM) showed increased viability and reduced ROS generation and apoptosis with GSH treatment (0.1 to 1 mM) for 24 h. In contrast to the 500 nM DOX group, pERK levels were restored in the group co-treated with GSH and suppression of ERK signaling improved hCPCs' survival. Similarly to the previous results, the reduced potency of hCPCs in the 100 nM DOX group, which did not affect cell viability, was ameliorated by co-treatment with GSH (0.1 to 1 mM). Furthermore, GSH was protected against DOX-induced cardiotoxicity in the in vivo model (DOX 20 mg/kg, GSH 100 mg/kg). These results suggest that GSH is a potential therapeutic strategy for DOX-induced cardiotoxicity, which performs its function via ROS reduction and pERK signal regulation.
Collapse
Affiliation(s)
- Eun Ji Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Woong Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jaewoo Choi
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hye Ji Lim
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sangmi Park
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Vinoth Kumar Rethineswaran
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jong Seong Ha
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jisoo Yun
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Young Joon Hong
- Department of Cardiology, Chonnam National University School of Medicine, Chonnam National University Hospital, Gwangju 61469, Republic of Korea;
| | - Young Jin Choi
- Department of Hemato-Oncology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
44
|
El-Horany HES, Atef MM, Abdel Ghafar MT, Fouda MH, Nasef NA, Hegab II, Helal DS, Elseady W, Hafez YM, Hagag RY, Seleem MA, Saleh MM, Radwan DA, Abd El-Lateef AE, Abd-Ellatif RN. Empagliflozin Ameliorates Bleomycin-Induced Pulmonary Fibrosis in Rats by Modulating Sesn2/AMPK/Nrf2 Signaling and Targeting Ferroptosis and Autophagy. Int J Mol Sci 2023; 24:ijms24119481. [PMID: 37298433 DOI: 10.3390/ijms24119481] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Pulmonary fibrosis (PF) is a life-threatening disorder that severely disrupts normal lung architecture and function, resulting in severe respiratory failure and death. It has no definite treatment. Empagliflozin (EMPA), a sodium-glucose cotransporter 2 (SGLT2) inhibitor, has protective potential in PF. However, the mechanisms underlying these effects require further elucidation. Therefore, this study aimed to evaluate the ameliorative effect of EMPA against bleomycin (BLM)-induced PF and the potential mechanisms. Twenty-four male Wister rats were randomly divided into four groups: control, BLM treated, EMPA treated, and EMPA+BLM treated. EMPA significantly improved the histopathological injuries illustrated by both hematoxylin and eosin and Masson's trichrome-stained lung tissue sections, as confirmed by electron microscopic examination. It significantly reduced the lung index, hydroxyproline content, and transforming growth factor β1 levels in the BLM rat model. It had an anti-inflammatory effect, as evidenced by a decrease in the inflammatory cytokines' tumor necrosis factor alpha and high mobility group box 1, inflammatory cell infiltration into the bronchoalveolar lavage fluid, and the CD68 immunoreaction. Furthermore, EMPA mitigated oxidative stress, DNA fragmentation, ferroptosis, and endoplasmic reticulum stress, as evidenced by the up-regulation of nuclear factor erythroid 2-related factor expression, heme oxygenase-1 activity, glutathione peroxidase 4 levels, and a decrease in C/EBP homologous protein levels. This protective potential could be explained on the basis of autophagy induction via up-regulating lung sestrin2 expression and the LC3 II immunoreaction observed in this study. Our findings indicated that EMPA protected against BLM-induced PF-associated cellular stress by enhancing autophagy and modulating sestrin2/adenosine monophosphate-activated protein kinase/nuclear factor erythroid 2-related factor 2/heme oxygenase 1 signaling.
Collapse
Affiliation(s)
- Hemat El-Sayed El-Horany
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
- Department of Biochemistry, College of Medicine, Ha'il University, Hail 81411, Saudi Arabia
| | - Marwa Mohamed Atef
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | | | - Mohamed H Fouda
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | - Nahla Anas Nasef
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | - Islam Ibrahim Hegab
- Physiology Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
- Department of Bio-Physiology, Ibn Sina National College for Medical Studies, Jeddah 22421, Saudi Arabia
| | - Duaa S Helal
- Pathology Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | - Walaa Elseady
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | - Yasser Mostafa Hafez
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | - Rasha Youssef Hagag
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | | | - Mai Mahmoud Saleh
- Chest Diseases Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | - Doaa A Radwan
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | | | - Rania Nagi Abd-Ellatif
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| |
Collapse
|
45
|
Hwang I, Kim M. Muscular Sestrins: Roles in Exercise Physiology and Stress Resistance. Biomolecules 2023; 13:722. [PMID: 37238592 PMCID: PMC10216764 DOI: 10.3390/biom13050722] [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: 03/21/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Sestrins are a family of stress-inducible proteins that are critical for stress adaptation and the maintenance of metabolic homeostasis. High expression of Sestrins is observed in skeletal and cardiac muscle tissues, suggesting their significance in the physiological homeostasis of these organs. Furthermore, expression of Sestrins is dynamically controlled in the tissues, based on the level of physical activity and the presence or absence of stress insults. Genetic studies in model organisms have shown that muscular Sestrin expression is critical for metabolic homeostasis, exercise adaptation, stress resistance, and repair and may mediate the beneficial effects of some available therapeutics. The current minireview summarizes and discusses recent findings that shed light on the role of Sestrins in regulating muscle physiology and homeostasis.
Collapse
Affiliation(s)
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
46
|
Bo L, Wang Y, Li Y, Wurpel JND, Huang Z, Chen ZS. The Battlefield of Chemotherapy in Pediatric Cancers. Cancers (Basel) 2023; 15:cancers15071963. [PMID: 37046624 PMCID: PMC10093214 DOI: 10.3390/cancers15071963] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
The survival rate for pediatric cancers has remarkably improved in recent years. Conventional chemotherapy plays a crucial role in treating pediatric cancers, especially in low- and middle-income countries where access to advanced treatments may be limited. The Food and Drug Administration (FDA) approved chemotherapy drugs that can be used in children have expanded, but patients still face numerous side effects from the treatment. In addition, multidrug resistance (MDR) continues to pose a major challenge in improving the survival rates for a significant number of patients. This review focuses on the severe side effects of pediatric chemotherapy, including doxorubicin-induced cardiotoxicity (DIC) and vincristine-induced peripheral neuropathy (VIPN). We also delve into the mechanisms of MDR in chemotherapy to the improve survival and reduce the toxicity of treatment. Additionally, the review focuses on various drug transporters found in common types of pediatric tumors, which could offer different therapeutic options.
Collapse
Affiliation(s)
- Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Youyou Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Yidong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - John N. D. Wurpel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Zoufang Huang
- Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Correspondence: (Z.H.); (Z.-S.C.); Tel.: +86-138-797-27439 (Z.H.); +1-718-990-1432 (Z.-S.C.); Fax: +1-718-990-1877 (Z.-S.C.)
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
- Institute for Biotechnology, St. John’s University, Queens, NY 11439, USA
- Correspondence: (Z.H.); (Z.-S.C.); Tel.: +86-138-797-27439 (Z.H.); +1-718-990-1432 (Z.-S.C.); Fax: +1-718-990-1877 (Z.-S.C.)
| |
Collapse
|
47
|
Kuang Z, Wu J, Tan Y, Zhu G, Li J, Wu M. MicroRNA in the Diagnosis and Treatment of Doxorubicin-Induced Cardiotoxicity. Biomolecules 2023; 13:biom13030568. [PMID: 36979503 PMCID: PMC10046787 DOI: 10.3390/biom13030568] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Doxorubicin (DOX), a broad-spectrum chemotherapy drug, is widely applied to the treatment of cancer; however, DOX-induced cardiotoxicity (DIC) limits its clinical therapeutic utility. However, it is difficult to monitor and detect DIC at an early stage using conventional detection methods. Thus, sensitive, accurate, and specific methods of diagnosis and treatment are important in clinical practice. MicroRNAs (miRNAs) belong to non-coding RNAs (ncRNAs) and are stable and easy to detect. Moreover, miRNAs are expected to become biomarkers and therapeutic targets for DIC; thus, there are currently many studies focusing on the role of miRNAs in DIC. In this review, we list the prominent studies on the diagnosis and treatment of miRNAs in DIC, explore the feasibility and difficulties of using miRNAs as diagnostic biomarkers and therapeutic targets, and provide recommendations for future research.
Collapse
Affiliation(s)
- Ziyu Kuang
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jingyuan Wu
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ying Tan
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guanghui Zhu
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jie Li
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Min Wu
- Cardiovascular Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
48
|
Yi X, Wang H, Yang Y, Wang H, Zhang H, Guo S, Chen J, Du J, Tian Y, Ma J, Zhang B, Wu L, Shi Q, Gao T, Guo W, Li C. SIRT7 orchestrates melanoma progression by simultaneously promoting cell survival and immune evasion via UPR activation. Signal Transduct Target Ther 2023; 8:107. [PMID: 36918544 PMCID: PMC10015075 DOI: 10.1038/s41392-023-01314-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/17/2022] [Accepted: 01/09/2023] [Indexed: 03/16/2023] Open
Abstract
Melanoma is the most lethal type of skin cancer, originating from the malignant transformation of melanocyte. While the development of targeted therapy and immunotherapy has gained revolutionary advances in potentiating the therapeutic effect, the prognosis of patients with melanoma is still suboptimal. During tumor progression, melanoma frequently encounters stress from both endogenous and exogenous sources in tumor microenvironment. SIRT7 is a nuclear-localized deacetylase of which the activity is highly dependent on intracellular nicotinamide adenine dinucleotide (NAD+), with versatile biological functions in maintaining cell homeostasis. Nevertheless, whether SIRT7 regulates tumor cell biology and tumor immunology in melanoma under stressful tumor microenvironment remains elusive. Herein, we reported that SIRT7 orchestrates melanoma progression by simultaneously promoting tumor cell survival and immune evasion via the activation of unfolded protein response. We first identified that SIRT7 expression was the most significantly increased one in sirtuins family upon stress. Then, we proved that the deficiency of SIRT7 potentiated tumor cell death under stress in vitro and suppressed melanoma growth in vivo. Mechanistically, SIRT7 selectively activated the IRE1α-XBP1 axis to potentiate the pro-survival ERK signal pathway and the secretion of tumor-promoting cytokines. SIRT7 directly de-acetylated SMAD4 to antagonize the TGF-β-SMAD4 signal, which relieved the transcriptional repression on IRE1α and induced the activation of the IRE1α-XBP1 axis. Moreover, SIRT7 up-regulation eradicated anti-tumor immunity by promoting PD-L1 expression via the IRE1α-XBP1 axis. Additionally, the synergized therapeutic effect of SIRT7 suppression and anti-PD-1 immune checkpoint blockade was also investigated. Taken together, SIRT7 can be employed as a promising target to restrain tumor growth and increase the effect of melanoma immunotherapy.
Collapse
Affiliation(s)
- Xiuli Yi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Yuqi Yang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Hao Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Hengxiang Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Sen Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Jianru Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Juan Du
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Yangzi Tian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Jingjing Ma
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Baolu Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Lili Wu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Qiong Shi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China.
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No 127 of West Changle Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
49
|
Li S, Huang Q, He B. SIRT1 as a Potential Therapeutic Target for Chronic Obstructive Pulmonary Disease. Lung 2023; 201:201-215. [PMID: 36790647 DOI: 10.1007/s00408-023-00607-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/04/2023] [Indexed: 02/16/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common, preventable, and treatable disease characterized by irreversible airflow obstruction and lung function decline. It is well established that COPD represents a major cause of morbidity and mortality globally. Due to the substantial economic and social burdens associated with COPD, it is necessary to discover new targets and develop novel beneficial therapies. Although the pathogenesis of COPD is complex and remains to be robustly elucidated, numerous studies have shown that oxidative stress, inflammatory responses, cell apoptosis, autophagy, and aging are involved in the pathogenesis of COPD. Sirtuin 1 (SIRT1) is a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase belonging to the silent information regulator 2 (Sir2) family. Multiple studies have indicated that SIRT1 plays an important role in oxidative stress, apoptosis, inflammation, autophagy, and cellular senescence, which contributes to the pathogenesis and development of COPD. This review aimed to discuss the functions and mechanisms of SIRT1 in the progression of COPD and concluded that SIRT1 activation might be a potential therapeutic strategy for COPD.
Collapse
Affiliation(s)
- Siqi Li
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.,Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Huang
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.,Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Baimei He
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China. .,Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
50
|
Kawano I, Adamcova M. MicroRNAs in doxorubicin-induced cardiotoxicity: The DNA damage response. Front Pharmacol 2022; 13:1055911. [PMID: 36479202 PMCID: PMC9720152 DOI: 10.3389/fphar.2022.1055911] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/11/2022] [Indexed: 10/17/2023] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic drug widely used for cancer treatment, but its use is limited by cardiotoxicity. Although free radicals from redox cycling and free cellular iron have been predominant as the suggested primary pathogenic mechanism, novel evidence has pointed to topoisomerase II inhibition and resultant genotoxic stress as the more fundamental mechanism. Recently, a growing list of microRNAs (miRNAs) has been implicated in DOX-induced cardiotoxicity (DIC). This review summarizes miRNAs reported in the recent literature in the context of DIC. A particular focus is given to miRNAs that regulate cellular responses downstream to DOX-induced DNA damage, especially p53 activation, pro-survival signaling pathway inhibition (e.g., AMPK, AKT, GATA-4, and sirtuin pathways), mitochondrial dysfunction, and ferroptosis. Since these pathways are potential targets for cardioprotection against DOX, an understanding of how miRNAs participate is necessary for developing future therapies.
Collapse
Affiliation(s)
| | - Michaela Adamcova
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University in Prague, Hradec Kralove, Czechia
| |
Collapse
|