1
|
Kim HJ, Kang SU, Kim HJ, Lee YS, Kim CH. GDF15 inhibits early-stage adipocyte differentiation by enhancing HOP2 expression and suppressing C/EBPα expression. Mol Cell Endocrinol 2025; 598:112461. [PMID: 39814165 DOI: 10.1016/j.mce.2025.112461] [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: 07/22/2024] [Revised: 01/02/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
Excessive adipocyte differentiation and accumulation contribute to the development of metabolic disorders. Growth differentiation factor 15 (GDF15) plays an essential role in energy homeostasis and is considered an anti-obesity factor; however, elevated serum levels of endogenous GDF15 have been reported in certain individuals with obesity. In this study, to gain a better understanding of this complex relationship between GDF15 levels and obesity, we investigated GDF15 expression and function during adipogenesis. Compared with mice fed a normal diet, those fed a short-term high-fat diet exhibited a reduction in epididymal white adipose tissue and serum GDF15 expression. These results were confirmed in human adipose-derived stem cells that showed reduced GDF15 expression during adipogenesis differentiation. During adipogenesis, GDF15 was primarily degraded via the autophagy lysosomal pathway, and GDF15 overexpression in pre-adipocytes inhibited adipogenesis by suppressing CCAAT enhancer binding protein alpha (C/EBPα). Furthermore, whereas we detected a reduction in homologous-pairing protein 2 (HOP2) expression during adipogenesis, expression increased in response to an overexpression of GDF15. Furthermore, following knockdown of HOP2 during GDF15 overexpression, there was no suppression of C/EBPα expression. These findings indicate that GDF15 undergoes lysosomal degradation via an autophagic pathway and suppresses adipocyte differentiation via the HOP2-mediated inhibition of C/EBPα expression. Collectively, our findings indicate that GDF15 could serve as a potential therapeutic target for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Haeng Jun Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Sung-Un Kang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Hyo Jeong Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Yun Sang Lee
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea.
| |
Collapse
|
2
|
Behera BP, Mishra SR, Patra S, Mahapatra KK, Bhol CS, Panigrahi DP, Praharaj PP, Klionsky DJ, Bhutia SK. Molecular regulation of mitophagy signaling in tumor microenvironment and its targeting for cancer therapy. Cytokine Growth Factor Rev 2025:S1359-6101(25)00004-8. [PMID: 39880721 DOI: 10.1016/j.cytogfr.2025.01.004] [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: 12/16/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
Aberrations emerging in mitochondrial homeostasis are restrained by mitophagy to control mitochondrial integrity, bioenergetics signaling, metabolism, oxidative stress, and apoptosis. The mitophagy-accompanied mitochondrial processes that occur in a dysregulated condition act as drivers for cancer occurrence. In addition, the enigmatic nature of mitophagy in cancer cells modulates the cellular proteome, creating challenges for therapeutic interventions. Several reports found the role of cellular signaling pathways in cancer to modulate mitophagy to mitigate stress, immune checkpoints, energy demand, and cell death. Thus, targeting mitophagy to hinder oncogenic intracellular signaling by promoting apoptosis, in hindsight, might have an edge against cancer. This review highlights the receptors and adaptors, and the involvement of many proteins in mitophagy and their role in oncogenesis. It also provides insight into using mitophagy as a potential target for therapeutic intervention in various cancer types.
Collapse
Affiliation(s)
- Bishnu Prasad Behera
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Soumya Ranjan Mishra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Kewal Kumar Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Chandra Sekhar Bhol
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Debasna Pritimanjari Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India.
| |
Collapse
|
3
|
Yun JH, Yang YH, Han CH, Kang SU, Kim CH. Non-thermal atmospheric pressure plasma induces selective cancer cell apoptosis by modulating redox homeostasis. Cell Commun Signal 2024; 22:452. [PMID: 39327567 PMCID: PMC11426099 DOI: 10.1186/s12964-024-01810-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Anticancer treatments aim to selectively target cancer cells without harming normal cells. While non-thermal atmospheric pressure plasma (NTAPP) has shown anticancer potential across various studies, the mechanisms behind its selective action on cancer cells remain inadequately understood. This study explores the mechanism of NTAPP-induced selective cell death and assesses its application in cancer therapy. METHODS We treated HT1080 fibrosarcoma cells with NTAPP and assessed the intracellular levels of mitochondria-derived reactive oxygen species (ROS), mitochondrial function, and cell death mechanisms. We employed N-acetylcysteine to investigate ROS's role in NTAPP-induced cell death. Additionally, single-cell RNA sequencing was used to compare gene expression in NTAPP-treated HT1080 cells and human normal fibroblasts (NF). Western blotting and immunofluorescence staining examined the expression and nuclear translocation of nuclear factor erythroid 2-related factor 2 (NRF2), a key antioxidant gene transcription factor. We also evaluated autophagy activity through fluorescence staining and transmission electron microscopy. RESULTS NTAPP treatment increased ROS levels and induced mitochondrial dysfunction, leading to apoptosis in HT1080 cells. The involvement of ROS in selective cancer cell death was confirmed by N-acetylcysteine treatment. Distinct gene expression patterns were observed between NTAPP-treated NF and HT1080 cells, with NF showing upregulated antioxidant gene expression. Notably, NRF2 expression and nuclear translocation increased in NF but not in HT1080 cells. Furthermore, autophagy activity was significantly higher in normal cells compared to cancer cells. CONCLUSIONS Our study demonstrates that NTAPP induces selective cell death in fibrosarcoma cells through the downregulation of the NRF2-induced ROS scavenger system and inhibition of autophagy. These findings suggest NTAPP's potential as a cancer therapy that minimizes damage to normal cells while effectively targeting cancer cells.
Collapse
Affiliation(s)
- Ju Hyun Yun
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, Seoul, Korea, 07985
| | - Yoon Hee Yang
- Department of Biomedical Sciences, Graduate School of Medicine, Ajou University, Suwon, Korea, 16499
| | - Chang Hak Han
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Korea, 16499
| | - Sung Un Kang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Korea, 16499.
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Korea, 16499.
| |
Collapse
|
4
|
Ulaganathan T, Perales S, Mani S, Baskhairoun BA, Rajasingh J. Pathological implications of cellular stress in cardiovascular diseases. Int J Biochem Cell Biol 2023; 158:106397. [PMID: 36931385 PMCID: PMC10124590 DOI: 10.1016/j.biocel.2023.106397] [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: 09/26/2022] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Cellular stress has been a key factor in the development of cardiovascular diseases. Major types of cellular stress such as mitochondrial stress, endoplasmic reticulum stress, hypoxia, and replicative stress have been implicated in clinical complications of cardiac patients. The heart is the central regulator of the body by supplying oxygenated blood throughout the system. Impairment of cellular function could lead to heart failure, myocardial infarction, ischemia, and even stroke. Understanding the effect of these distinct types of cellular stress on cardiac function is crucial for the scientific community to understand and develop novel therapeutic approaches. This review will comprehensively explain the different mechanisms of cellular stress and the most recent findings related to stress-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Thennavan Ulaganathan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Biotechnology, SRM Institute of Science and Technology, kattankulathur, Tamilnadu, 603203, India
| | - Selene Perales
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Saiprahalad Mani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Biotechnology, SRM Institute of Science and Technology, kattankulathur, Tamilnadu, 603203, India
| | - Boula A Baskhairoun
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Medicine-Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
5
|
Tang Y, Zhang J, Hu Z, Xu W, Xu P, Ma Y, Xing H, Niu Q. PRKAA1 induces aberrant mitophagy in a PINK1/Parkin-dependent manner, contributing to fluoride-induced developmental neurotoxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 255:114772. [PMID: 36924562 DOI: 10.1016/j.ecoenv.2023.114772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/02/2023] [Accepted: 03/11/2023] [Indexed: 06/18/2023]
Abstract
Chronic fluoride exposure can cause developmental neurotoxicity, however the precise mechanisms remain unclear. To explore the mechanism of mitophagy in fluoride-induced developmental neurotoxicity, specifically focusing on PRKAA1 in regulating the PINK1/Parkin pathway, we established a Sprage Dawley rat model with continuous sodium fluoride (NaF) exposure and an NaF-treated SH-SY5Y cell model. We found that NaF exposure increased the levels of LC3-Ⅱ and p62, impaired autophagic degradation, and subsequently blocked autophagic flux. Additionally, NaF exposure increased the expression of PINK1, Parkin, TOMM-20, and Cyt C and cleaved PARP in vivo and in vitro, indicating NaF promotes mitophagy and neuronal apoptosis. Meanwhile, phosphoproteomics and western blot analysis showed that NaF treatment enhanced PRKAA1 phosphorylation. Remarkably, the application of both 3-methyladenosine (3-MA; autophagy inhibitor) and dorsomorphin (DM; AMPK inhibitor) suppressed NaF-induced neuronal apoptosis by restoring aberrant mitophagy. In addition, 3-MA attenuated an increase in p62 protein levels and NaF-induced autophagic degradation. Collectively, our findings indicated that NaF causes aberrant mitophagy via PRKAA1 in a PINK1/Parkin-dependent manner, which triggers neuronal apoptosis. Thus, regulating PRKAA1-activated PINK1/Parkin-dependent mitophagy may be a potential treatment for NaF-induced developmental neurotoxicity.
Collapse
Affiliation(s)
- Yanling Tang
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), People's Republic of China
| | - Jingjing Zhang
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), People's Republic of China
| | - Zeyu Hu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), People's Republic of China
| | - Wanjing Xu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), People's Republic of China
| | - Panpan Xu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), People's Republic of China
| | - Yue Ma
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), People's Republic of China
| | - Hengrui Xing
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), People's Republic of China
| | - Qiang Niu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), People's Republic of China.
| |
Collapse
|
6
|
Lee HR, Kang SU, Kim HJ, Ji EJ, Yun JH, Kim S, Jang JY, Shin YS, Kim CH. Liquid plasma as a treatment for cutaneous wound healing through regulation of redox metabolism. Cell Death Dis 2023; 14:119. [PMID: 36781835 PMCID: PMC9925775 DOI: 10.1038/s41419-023-05610-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 02/15/2023]
Abstract
The skin functions as the outermost protective barrier to the internal organs and major vessels; thus, delayed regeneration from acute injury could induce serious clinical complications. For rapid recovery of skin wounds, promoting re-epithelialization of the epidermis at the initial stage of injury is essential, wherein epithelial keratinocytes act as leading cells via migration. This study applied plasma technology, which has been known to enable wound healing in the medical field. Through in vitro and in vivo experiments, the study elucidated the effect and molecular mechanism of the liquid plasma (LP) manufactured by our microwave plasma system, which was found to improve the applicability of existing gas-type plasma on skin cell migration for re-epithelialization. LP treatment promoted the cytoskeletal transformation of keratinocytes and migration owing to changes in the expression of integrin-dependent focal adhesion molecules and matrix metalloproteinases (MMPs). This study also identified the role of increased levels of intracellular reactive oxygen species (ROS) as a driving force for cell migration activation, which was regulated by changes in NADPH oxidases and mitochondrial membrane potential. In an in vivo experiment using a murine dorsal full-thickness acute skin wound model, LP treatment helped improve the re-epithelialization rate, reaffirming the activation of the underlying intracellular ROS-dependent integrin-dependent signaling molecules. These findings indicate that LP could be a valuable wound management material that can improve the regeneration potential of the skin via the activation of migration-related molecular signaling within the epithelial cell itself with plasma-driven oxidative eustress.
Collapse
Affiliation(s)
- Hye Ran Lee
- Department of Otolaryngology-Head and Neck Surgery, Catholic Kwandong University International St. Mary's Hospital, Incheon, 22711, Republic of Korea
- Department of Medical Sciences, Otolaryngology, Graduate School of Ajou University, Suwon, 16499, Republic of Korea
| | - Sung Un Kang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Haeng Jun Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Eun Jong Ji
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Ju Hyun Yun
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Sungryeal Kim
- Department of Otolaryngology, College of Medicine, Inha University, Incheon, 22332, Republic of Korea
| | - Jeon Yeob Jang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Yoo Seob Shin
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea.
| |
Collapse
|
7
|
Ma N, Shangguan F, Zhou H, Huang H, Lei J, An J, Jin G, Zhuang W, Zhou S, Wu S, Xia H, Yang H, Lan L. 6-methoxydihydroavicine, the alkaloid extracted from Macleaya cordata (Willd.) R. Br. (Papaveraceae), triggers RIPK1/Caspase-dependent cell death in pancreatic cancer cells through the disruption of oxaloacetic acid metabolism and accumulation of reactive oxygen species. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 102:154164. [PMID: 35597026 DOI: 10.1016/j.phymed.2022.154164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Many extracts and purified alkaloids of M. cordata (Papaveraceae family) have been reported to display promising anti-tumor effects by inhibiting cancer cell growth and inducing apoptosis in many cancer types. However, no evidence currently exists for anti-pancreatic cancer activity of alkaloids extracted from M. cordata, including a novel alkaloid named 6‑methoxy dihydrosphingosine (6-Methoxydihydroavicine, 6-ME) derived from M. cordata fruits. PURPOSE The aim of this study was to investigate the anti-tumor effects of 6-ME on PC cells and the underlying mechanism. METHODS CCK-8, RTCA, and colony-formation assays were used to analyze PC cell growth. Cell death ratios, changes in MMP and ROS levels were measured by flow cytometry within corresponding detection kits. A Seahorse XFe96 was employed to examine the effects of 6-ME on cellular bioenergetics. Western blot and q-RT-PCR were conducted to detect changes in target molecules. RESULTS 6-ME effectively reduced the growth of PC cells and promoted PCD by activating RIPK1, caspases, and GSDME. Specifically, 6-ME treatment caused a disruption of OAA metabolism and increased ROS production, thereby affecting mitochondrial homeostasis and reducing aerobic glycolysis. These responses resulted in mitophagy and RIPK1-mediated cell death. CONCLUSION 6-ME exhibited specific anti-tumor effects through interrupting OAA metabolic homeostasis to trigger ROS/RIPK1-dependent cell death and mitochondrial dysfunction, suggesting that 6-ME could be considered as a highly promising compound for PC intervention.
Collapse
Affiliation(s)
- Nengfang Ma
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou 325000, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Fugen Shangguan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Hongfei Zhou
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Huimin Huang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University Town, Ouhai District, Wenzhou 325000, China
| | - Jun Lei
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, Department of Biochemistry and Molecular Biology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jing An
- Division of Infectious Diseases and Global Health, School of Medicine, University of California San Diego (UCSD), LaJolla, CA 92037, United States of America
| | - Guihua Jin
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Weiwei Zhuang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Shipeng Zhou
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Shijia Wu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Hongping Xia
- Henan Medical School & Huaihe Hospital & The First Affiliated Hospital, Henan University, Kaifeng, China.
| | - Hailong Yang
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou 325000, China.
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
8
|
Lu J, Fu S, Dai J, Hu J, Li S, Ji H, Wang Z, Yu J, Bao J, Xu B, Guo J, Yang H. Integrated metabolism and epigenetic modifications in the macrophages of mice in responses to cold stress. J Zhejiang Univ Sci B 2022; 23:461-480. [PMID: 35686526 PMCID: PMC9198231 DOI: 10.1631/jzus.b2101091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The negative effects of low temperature can readily induce a variety of diseases. We sought to understand the reasons why cold stress induces disease by studying the mechanisms of fine-tuning in macrophages following cold exposure. We found that cold stress triggers increased macrophage activation accompanied by metabolic reprogramming of aerobic glycolysis. The discovery, by genome-wide RNA sequencing, of defective mitochondria in mice macrophages following cold exposure indicated that mitochondrial defects may contribute to this process. In addition, changes in metabolism drive the differentiation of macrophages by affecting histone modifications. Finally, we showed that histone acetylation and lactylation are modulators of macrophage differentiation following cold exposure. Collectively, metabolism-related epigenetic modifications are essential for the differentiation of macrophages in cold-stressed mice, and the regulation of metabolism may be crucial for alleviating the harm induced by cold stress.
Collapse
Affiliation(s)
- Jingjing Lu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Shoupeng Fu
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jie Dai
- Shanghai Bioprofile Co. Ltd., Shanghai 201100, China
| | - Jianwen Hu
- Shanghai Bioprofile Co. Ltd., Shanghai 201100, China
| | - Shize Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Hong Ji
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Zhiquan Wang
- Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Alberta T5J 4P6, Canada
| | - Jiahong Yu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jiming Bao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Bin Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jingru Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| | - Huanmin Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| |
Collapse
|
9
|
Fang Z, Li X, Wang S, Jiang Q, Loor JJ, Jiang X, Ju L, Yu H, Shen T, Chen M, Song Y, Wang Z, Du X, Liu G. Overactivation of hepatic mechanistic target of rapamycin kinase complex 1 (mTORC1) is associated with low transcriptional activity of transcription factor EB and lysosomal dysfunction in dairy cows with clinical ketosis. J Dairy Sci 2022; 105:4520-4533. [DOI: 10.3168/jds.2021-20892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 01/14/2022] [Indexed: 11/19/2022]
|
10
|
Zhao Y, Li HX, Luo Y, Cui JG, Talukder M, Li JL. Lycopene mitigates DEHP-induced hepatic mitochondrial quality control disorder via regulating SIRT1/PINK1/mitophagy axis and mitochondrial unfolded protein response. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 292:118390. [PMID: 34699919 DOI: 10.1016/j.envpol.2021.118390] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/25/2021] [Accepted: 10/18/2021] [Indexed: 06/13/2023]
Abstract
Di (2-ethylhexyl) phthalate (DEHP) is a hazardous chemical which is used as a plasticizer in the plastic products. Lycopene (LYC) is a carotenoid that has protective roles against cellular damage in different organs. The present study sought to explore the role of the interaction between mitophagy and mitochondrial unfolded protein response (UPRmt) in the LYC mitigating DEHP-induced hepatic mitochondrial quality control disorder. The mice were treated with LYC (5 mg/kg) and/or DEHP (500 or 1000 mg/kg). In our findings, LYC prevented DEHP-induced histopathological alterations including steatosis and fibrosis, and ultrastructural injuries including decreased mitochondrial membrane potential (ΔΨm) and mitochondria volume density. Furthermore, LYC alleviated DEHP-induced mitochondrial biogenesis disorder by suppressing SIRT1-PGC-1α axis, PINK1-mediated mitophagy and the activation of mitochondrial unfolded protein response (UPRmt). This research suggested that LYC could prevent DEHP-induced hepatic mitochondrial quality control disorder via regulating SIRT1/PINK1/mitophagy axis and UPRmt. The present study provided a current understanding about the potential implication of the SIRT1/PINK1/mitophagy axis and UPRmt in LYC preventing DEHP-induced hepatic mitochondrial quality control disorder.
Collapse
Affiliation(s)
- Yi Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Hui-Xin Li
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150086, PR China
| | - Yu Luo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; People's Government of Nierhe Township of Suiling County, Suihua, 152236, PR China
| | - Jia-Gen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Milton Talukder
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, 8210, Bangladesh
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
11
|
Lee HR, Lee HY, Heo J, Jang JY, Shin YS, Kim CH. Liquid-type nonthermal atmospheric plasma enhanced regenerative potential of silk-fibrin composite gel in radiation-induced wound failure. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112304. [PMID: 34474855 DOI: 10.1016/j.msec.2021.112304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 06/29/2021] [Accepted: 07/04/2021] [Indexed: 12/15/2022]
Abstract
Delayed wound healing in heavily irradiated areas is a serious clinical complication that makes widespread therapeutic use of radiation difficult. Efficient treatment strategies are urgently required for addressing radiation-induced wound failure. Herein, we applied liquid-type nonthermal atmospheric plasma (LTP) to a silk-fibrin (SF) composite gel to investigate whether controlled release of LTP from SF hydrogel not only induced favorable cellular events in an irradiated wound bed but also modulated the SF hydrogel microstructure itself, eventually facilitating the development of a regenerative microenvironment. Scanning electron microscopy and Fourier-transform infrared spectroscopy revealed that LTP modulated the microstructures and chemical bindings of the SF gel. Improved cell viability, morphology, and extracellular matrix depositions by the LTP-treated SF hydrogel were identified with wound-healing assays and immunofluorescence staining. An irradiated random-pattern skin-flap animal model was established in six-week-old C57/BL6 mice. Full-thickness skin was flapped from the dorsum and SF hydrogel was placed underneath the raised skin flap. Postoperative histological analysis of the irradiated random-pattern skin-flap mice model suggested that LTP-treated SF hydrogel much improved wound regeneration and the inflammatory response compared to the SF hydrogel- and sham-treated groups. These results support that LTP-treated SF hydrogel significantly enhanced irradiated wound healing. Cellular and tissue reactions to released LTP from the SF hydrogel were favorable for the regenerative process of the wound; furthermore, mechanochemical properties of the SF gel were improved by LTP.
Collapse
Affiliation(s)
- Hye Ran Lee
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Hye-Young Lee
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Jaesung Heo
- Department of Radiation Oncology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Jeon Yeob Jang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Yoo Seob Shin
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea.
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| |
Collapse
|