1
|
Wang K, Zhang L, Deng B, Zhao K, Chen C, Wang W. Mitochondrial uncoupling protein 2: a central player in pancreatic disease pathophysiology. Mol Med 2024; 30:259. [PMID: 39707176 DOI: 10.1186/s10020-024-01027-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
Pancreatic diseases pose considerable health challenges due to their complex etiology and limited therapeutic options. Mitochondrial uncoupling protein 2 (UCP2), highly expressed in pancreatic tissue, participates in numerous physiological processes and signaling pathways, indicating its potential relevance in these diseases. Despite this, UCP2's role in acute pancreatitis (AP) remains underexplored, and its functions in chronic pancreatitis (CP) and pancreatic steatosis are largely unknown. Additionally, the mechanisms connecting various pancreatic diseases are intricate and not yet fully elucidated. Given UCP2's diverse functionality, broad expression in pancreatic tissue, and the distinct pathophysiological features of pancreatic diseases, this review offers a comprehensive analysis of current findings on UCP2's involvement in these conditions. We discuss recent insights into UCP2's complex regulatory mechanisms, propose that UCP2 may serve as a central regulatory factor in pancreatic disease progression, and hypothesize that UCP2 dysfunction could significantly contribute to disease pathogenesis. Understanding UCP2's role and mechanisms in pancreatic diseases may pave the way for innovative therapeutic and diagnostic approaches.
Collapse
Affiliation(s)
- Kunpeng Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Beiying Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kailiang Zhao
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Tan R, Ge C, Yan Y, Guo H, Han X, Zhu Q, Du Q. Deciphering ferroptosis in critical care: mechanisms, consequences, and therapeutic opportunities. Front Immunol 2024; 15:1511015. [PMID: 39737174 PMCID: PMC11682965 DOI: 10.3389/fimmu.2024.1511015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/03/2024] [Indexed: 01/01/2025] Open
Abstract
Ischemia-reperfusion injuries (IRI) across various organs and tissues, along with sepsis, significantly contribute to the progression of critical illnesses. These conditions disrupt the balance of inflammatory mediators and signaling pathways, resulting in impaired physiological functions in human tissues and organs. Ferroptosis, a distinct form of programmed cell death, plays a pivotal role in regulating tissue damage and modulating inflammatory responses, thereby influencing the onset and progression of severe illnesses. Recent studies highlight that pharmacological agents targeting ferroptosis-related proteins can effectively mitigate oxidative stress caused by IRI in multiple organs, alleviating associated symptoms. This manuscript delves into the mechanisms and signaling pathways underlying ferroptosis, its role in critical illnesses, and its therapeutic potential in mitigating disease progression. We aim to offer a novel perspective for advancing clinical treatments for critical illnesses.
Collapse
Affiliation(s)
- Ruimin Tan
- School of Clinical Medical, North China University of Science and Technology, Tangshan, Hebei, China
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Chen Ge
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yating Yan
- School of Clinical Medical, North China University of Science and Technology, Tangshan, Hebei, China
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - He Guo
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
- School of Graduate, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xumin Han
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
- School of Graduate, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiong Zhu
- Department of Orthopaedics, The People’s Hospital Of Shizhu, Chongqing, China
| | - Quansheng Du
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
3
|
Wang H, Chen X, Hu D, Xin X, Zhao Z, Jiang Z. Reduced glutathione attenuates pediatric sepsis-associated encephalopathy by inhibiting inflammatory cytokine release and mitigating lipid peroxidation-induced brain injury. Neuroreport 2024; 35:1143-1154. [PMID: 39445523 DOI: 10.1097/wnr.0000000000002109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Sepsis-associated encephalopathy (SAE) is a severe complication of sepsis. Reduced glutathione (GSH) has antioxidant properties and is used as a neuroprotective agent in some studies. However, research on the application of exogenous GSH in the treatment of SAE is limited. This study aimed to determine the effects of exogenous GSH in pediatric SAE patients and mice. We evaluated clinical parameters, inflammatory factors, and oxidative stress before and after GSH treatment. The clinical trials demonstrated that GSH treatment improved brain damage markers (S-100 beta protein, brain fatty acid-binding protein), increased neurological status scores (Glasgow coma scale), and reduced Pediatric Risk of Mortality III scores in children with SAE. GSH treatment also significantly reduced the levels of inflammatory factors (interleukin-6, tumor necrosis factor-α) and decreased lipid peroxidation (superoxide dismutase). Additionally, GSH reduced lipid peroxidation resulting from abnormal lipid metabolism, as indicated by the levels of acyl-CoA synthetase long-chain family member 4, lysophosphatidylcholine acyltransferase 3, and glutathione peroxidase 4. In-vivo experiments showed that the neuroprotective effect of GSH was dose-dependent, with better effects observed at medium and high doses. Furthermore, GSH alleviated brain damage, suppressed the release of inflammatory factors, and inhibited lipid peroxidation in SAE mice. The animal experiments also showed that GSH reduces lipid peroxidation through the 15-lipoxygenase/phosphatidylethanolamine binding protein 1/glutathione peroxidase 4 pathway. Our study suggests that exogenous GSH has neuroprotective effects in pediatric SAE. These findings provide a basis for the potential use of GSH as a therapeutic method for SAE.
Collapse
Affiliation(s)
- Haosen Wang
- Department of Critical Care Medicine, The Affiliated Xuzhou Children℉s Hospital of Xuzhou Medical University
| | - Xinrui Chen
- Department of Pediatrics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dan Hu
- Department of Critical Care Medicine, The Affiliated Xuzhou Children℉s Hospital of Xuzhou Medical University
| | - Xin Xin
- Department of Critical Care Medicine, The Affiliated Xuzhou Children℉s Hospital of Xuzhou Medical University
| | - Zhongxiu Zhao
- Department of Critical Care Medicine, The Affiliated Xuzhou Children℉s Hospital of Xuzhou Medical University
| | - Zhen Jiang
- Department of Critical Care Medicine, The Affiliated Xuzhou Children℉s Hospital of Xuzhou Medical University
| |
Collapse
|
4
|
Lu Z, Wang X, Lin X, Mostafa S, Zou H, Wang L, Jin B. Plant anthocyanins: Classification, biosynthesis, regulation, bioactivity, and health benefits. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2024; 217:109268. [PMID: 39520908 DOI: 10.1016/j.plaphy.2024.109268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/27/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Anthocyanins are naturally water-soluble pigments of plants, which can be pink, orange, red, purple, or blue. Anthocyanins belong to a subcategory of flavonoids known as polyphenols and are consumed in plant-based foods. The antioxidant properties of anthocyanins benefit human health. However, there has been no comprehensive review of the classification, distribution, and biosynthesis of anthocyanins and their regulation in plants, along with their potential health benefits. In this review, we provide a systematic synthesis of recent progress in anthocyanin research, specifically focusing on the classification, biosynthetic pathways, regulatory mechanisms, bioactivity, and health benefits. We bridge the gaps in understanding anthocyanin biological significance and potential applications. Furthermore, we discuss future directions for anthocyanin research, such as biotechnology, bioavailability, and the integration of artificial intelligence. We highlight pivotal research questions that warrant further exploration in the field of anthocyanin research.
Collapse
Affiliation(s)
- Zhaogeng Lu
- College of Horticulture and Landscape, Yangzhou University, Yangzhou, 225009, China
| | - Xinwen Wang
- College of Horticulture and Landscape, Yangzhou University, Yangzhou, 225009, China
| | - Xinyi Lin
- College of Horticulture and Landscape, Yangzhou University, Yangzhou, 225009, China
| | - Salma Mostafa
- College of Horticulture and Landscape, Yangzhou University, Yangzhou, 225009, China
| | - Helin Zou
- College of Horticulture and Landscape, Yangzhou University, Yangzhou, 225009, China
| | - Li Wang
- College of Horticulture and Landscape, Yangzhou University, Yangzhou, 225009, China
| | - Biao Jin
- College of Horticulture and Landscape, Yangzhou University, Yangzhou, 225009, China; Key Laboratory of Plant Functional Genomics of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
5
|
Sood R, Sanjay, Kang SU, Yoon NY, Lee HJ. Malvidin-3-O-Glucoside Mitigates α-Syn and MPTP Co-Induced Oxidative Stress and Apoptosis in Human Microglial HMC3 Cells. Int J Mol Sci 2024; 25:12733. [PMID: 39684444 DOI: 10.3390/ijms252312733] [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: 09/11/2024] [Revised: 11/08/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Parkinson's disease (PD) is a widespread age-related neurodegenerative disorder characterized by the presence of an aggregated protein, α-synuclein (α-syn), which is encoded by the SNCA gene and localized to presynaptic terminals in a normal human brain. The α-syn aggregation is induced by the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mitochondrial neurotoxin and is therefore used to mimic PD-like pathology in various in vitro and in vivo models. However, in vitro PD-like pathology using α-syn and MPTP in human microglial cells has not yet been reported. Malvidin-3-O-glucoside (M3G) is a major anthocyanin primarily responsible for pigmentation in various fruits and beverages and has been reported to possess various bioactivities. However, the neuroprotective effects of M3G in humanized in vitro PD-like pathologies have not been reported. Therefore, individual and co-treatments of α-syn and MPTP in a human microglial (HMC3) cell line were used to establish a humanized PD-like pathology model in vitro. The individual treatments were significantly less cytotoxic when compared to the α-syn and MPTP co-treatment. This study examined the neuroprotective effects of M3G by treating HMC3 cells with α-syn (8 μg/mL) and MPTP (2 mM) individually or in a co-treatment in the presence or absence of M3G (50 μM). M3G demonstrated anti-apoptotic, anti-inflammatory, and antioxidative properties against the α-syn- and MPTP-generated humanized in vitro PD-like pathology. This study determined that the cytoprotective effects of M3G are mediated by nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase (HO)-1 signaling.
Collapse
Affiliation(s)
- Rachit Sood
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Republic of Korea
| | - Sanjay
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Republic of Korea
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Na Young Yoon
- Food Safety and Processing Research Division, National Institute of Fisheries Science, Busan 46083, Republic of Korea
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Science and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
6
|
Dong H, Sun K, Wang X, Cui M, Ma Y, Li K, Duan W, Zhang H, Zhang L, Sheng Z, He M, Zhang B. Repurposed genipin targeting UCP2 exhibits antitumor activity through inducing ferroptosis in glioblastoma. Acta Biochim Biophys Sin (Shanghai) 2024. [PMID: 39523775 DOI: 10.3724/abbs.2024168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Uncoupling protein-2 (UCP2) controls the antioxidant response and redox homeostasis in cancer and is considered a potent molecular target for cancer treatment. However, the specific mechanism of UCP2 inhibition and its role in glioblastoma (GBM) have not yet been elucidated. Here, we attempt to identify a UCP2 inhibitor and study the underlying molecular mechanism in GBM. Bioinformatics analysis and immunohistochemistry are used to validate the high expression of UCP2 in GBM and its prognostic significance. Drug intervention and tumor xenograft experiments are conducted to determine the inhibitory effect of genipin, a UCP2 inhibitor, on UCP2. The mitochondrial membrane potential and key ferroptosis genes are examined to determine the occurrence of ferroptosis. High expression of UCP2 in GBM is associated with poor prognosis, and inhibiting UCP2 can alleviate the malignant behavior of GBM tumors. Genipin can downregulate the expression of GPX4 and upregulate the expression of ACSL4 by inhibiting UCP2, leading to ferroptosis and alleviating the malignant behavior of tumors. In summary, UCP2 is a potential therapeutic target for GBM. Genipin, which targets UCP2, effectively inhibits GBM development by inducing ferroptosis in vivo and in vitro. These findings indicate that genipin treatment based on UCP2 targeting has potential therapeutic applications with a clinical perspective for the treatment of GBM patients.
Collapse
Affiliation(s)
- Hao Dong
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
| | - Kaixuan Sun
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang 261041, China
| | - Xuejie Wang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
| | - Meimei Cui
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
| | - Yaping Ma
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
| | - Kexin Li
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
| | - Wanli Duan
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
| | - Hongxing Zhang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
| | - Liying Zhang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
| | - Zhimei Sheng
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
| | - Maotao He
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang 261041, China
| | - Baogang Zhang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261042, China
| |
Collapse
|
7
|
Guo J, Kong D, Luo J, Xiong T, Wang F, Deng M, Kong Z, Yang S, Da J, Chen C, Lan J, Chu L, Han G, Liu J, Tan Y, Zhang J. Orexin-A Attenuates the Inflammatory Response in Sepsis-Associated Encephalopathy by Modulating Oxidative Stress and Inhibiting the ERK/NF-κB Signaling Pathway in Microglia and Astrocytes. CNS Neurosci Ther 2024; 30:e70096. [PMID: 39508266 PMCID: PMC11541240 DOI: 10.1111/cns.70096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Oxidative stress-induced inflammation is a major pathogenic mechanism in sepsis-associated encephalopathy (SAE). We hypothesized that regulation of reactive oxygen species (ROS) by the neuropeptide orexin-A could prevent SAE-induced oxidative stress and inflammation. Therefore, the aim of this study was to investigate the effects of orexin-A on oxidative stress and inflammation in SAE in mice. METHODS Adult male mice were treated with orexin-A (250 μg/kg, intranasal administration) to establish a cecal ligation perforation (CLP) model. We performed behavioral tests, observed neuronal damage in the hippocampal region, measured the levels of ROS, NOX2, and observed the structure of mitochondria by transmission electron microscopy. We then examined the inflammatory factors TNF-α and IL-1β, the activation of microglia and astrocytes, the expression of ERK/NF-κB, C3, and S100A10, and the presence of A1 type astrocytes and A2 type astrocytes. RESULTS Orexin-A treatment improved cognitive performance in CLP-induced SAE mice, attenuated neuronal apoptosis in the hippocampal region, ameliorated ROS levels and the extent of mitochondrial damage, and reduced protein expression of NOX2 in hippocampal tissue. In addition, orexin-A treatment significantly reduced microglia and astrocyte activation, inhibited the levels of P-ERK and NF-κB, and reduced the release of IL-1β and TNF-α, which were significantly increased after CLP. Finally, Orexin-A treatment significantly decreased the number of C3/glial fibrillary acidic protein (GFAP)-positive cells and increased the number of S100A10/GFAP-positive cells. CONCLUSION Our data suggest that orexin-A reduces ROS expression by inhibiting CLP-induced NOX2 production, thereby attenuating mitochondrial damage and neuronal apoptosis. Its inhibition of microglial and A1-type astrocyte activation and inflammation was associated with the ERK/NF-κB pathway. These suggest that orexin-A may reduce cognitive impairment in SAE by reducing oxidative stress-induced inflammation.
Collapse
Affiliation(s)
- Jing Guo
- GuiZhou University Medical CollegeGuiyangGuizhouChina
| | | | - Junchi Luo
- Department of NeurosurgeryGuizhou Provincial People's HospitalGuiyangChina
| | - Tao Xiong
- Department of NeurosurgeryGuizhou Provincial People's HospitalGuiyangChina
| | - Fang Wang
- GuiZhou University Medical CollegeGuiyangGuizhouChina
| | - Mei Deng
- Department of NeurosurgeryGuizhou Provincial People's HospitalGuiyangChina
| | - Zhuo Kong
- Department of NeurosurgeryGuizhou Provincial People's HospitalGuiyangChina
| | - Sha Yang
- GuiZhou University Medical CollegeGuiyangGuizhouChina
| | - Jingjing Da
- Department of NephrologyGuizhou Provincial People's HospitalGuiyangChina
| | - Chaofei Chen
- Institute of Pediatrics, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Jinhai Lan
- Department of the Second SurgeryZiyun People's HospitalAnshunChina
| | - Liangzhao Chu
- Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Guoqiang Han
- Department of NeurosurgeryGuizhou Provincial People's HospitalGuiyangChina
| | - Jian Liu
- Department of NeurosurgeryGuizhou Provincial People's HospitalGuiyangChina
| | - Ying Tan
- Department of NeurosurgeryGuizhou Provincial People's HospitalGuiyangChina
| | - Jiqin Zhang
- Department of AnesthesiologyGuizhou Provincial People's HospitalGuiyangChina
| |
Collapse
|
8
|
Zhao Q, Liu G, Ding Q, Zheng F, Shi X, Lin Z, Liang Y. The ROS/TXNIP/NLRP3 pathway mediates LPS-induced microglial inflammatory response. Cytokine 2024; 181:156677. [PMID: 38896955 DOI: 10.1016/j.cyto.2024.156677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is a diffuse brain dysfunction activated by microglia. The potential pathological changes of SAE are complex, and the cellular pathophysiological characteristics remains unclear. This study aims to explore the ROS/TXNIP/NLRP3 pathway mediated lipopolysaccharide (LPS)-induced inflammatory response in microglia. METHODS BV-2 cells were pre-incubated with 10 μM N-acetyl-L-cysteine (NAC) for 2 h, which were then reacted with 1 μg/mL LPS for 24 h. Western blot assay examined the protein levels of IBA1, CD68, TXNIP, NLRP3, ASC, and Cleaved Caspase-1 in BV-2 cells. The contents of inflammatory factor were detected by ELISA assay. The co-immunoprecipitation assay examined the interaction between TXNIP and NLRP3. RESULTS LPS was confirmed to promote the positive expressions of IBA1 and CD68 in BV-2 cells. The further experiments indicated that LPS enhanced ROS production and NLRP3 inflammasome activation in BV-2 cells. Moreover, we also found that NAC partially reversed the facilitation of LPS on the levels of ROS, IL-1β, IL-18, TXNIP, NLRP3, ASC, and Cleaved Caspase-1 in BV-2 cells. NAC treatment also notably alleviated the interaction between TXNIP and NLRP3 in BV-2 cells. CONCLUSION ROS inhibition mediated NLRP3 signaling inactivation by decreasing TXNIP expression.
Collapse
Affiliation(s)
- Qianlei Zhao
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, PR China
| | - Guanhao Liu
- Wenzhou Medical University, Wenzhou, Zhejiang 325027, PR China
| | - Qiang Ding
- Wenzhou Medical University, Wenzhou, Zhejiang 325027, PR China
| | - Feixia Zheng
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, PR China
| | - Xulai Shi
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, PR China
| | - Zhongdong Lin
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, PR China
| | - Yafeng Liang
- Department of Pediatric Emergency, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, PR China.
| |
Collapse
|
9
|
Bu Z, Xu S, Xu F. Deciphering the mechanism of cimifugin in mitigating LPS-induced neuroinflammation in BV-2 cells. Allergol Immunopathol (Madr) 2024; 52:38-45. [PMID: 38970263 DOI: 10.15586/aei.v52i4.1107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 04/29/2024] [Indexed: 07/08/2024]
Abstract
PURPOSE Sepsis often triggers a systemic inflammatory response leading to multi-organ dysfunction, with complex and not fully understood pathogenesis. This study investigates the therapeutic effects of cimifugin on BV-2 cells under sepsis-induced stress conditions. METHODS We utilized a BV-2 microglial cell model treated with lipopolysaccharide (LPS) to mimic sepsis. Assessments included cellular vitality, inflammatory cytokine quantification (6 interleukin [6IL]-1β, interleukin 6 [IL-6], and tumor necrosis factor-α [TNF-α]) via enzyme-linked-immunosorbent serologic assay, and analysis of mRNA expression using real-time polymerase chain reaction. Oxidative stress and mitochondrial function were also evaluated to understand the cellular effects of cimifugin. RESULTS Cimifugin significantly attenuated LPS-induced inflammatory responses, oxidative stress, and mitochondrial dysfunction. It enhanced cell viability and modulated the secretion and gene expression of inflammatory cytokines IL-1β, IL-6, and TNF-α. Notably, cimifugin activated the deacetylase sirtuin 1-nuclear factor erythroid 2-related factor 2 pathway, contributing to its protective effects against mitochondrial damage. CONCLUSION Cimifugin demonstrates the potential of being an effective treatment for sepsis--induced neuroinflammation, warranting further investigation.
Collapse
Affiliation(s)
- Zhang Bu
- Department of Emergency Medicine, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shan Xu
- Soochow University Campus Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Feng Xu
- Department of Emergency Medicine, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China;
| |
Collapse
|
10
|
Li X, Lin Z, Xu S, Zhang N, Zhou J, Liao B. Knockdown of KBTBD7 attenuates septic lung injury by inhibiting ferroptosis and improving mitochondrial dysfunction. Int Immunopharmacol 2024; 133:112129. [PMID: 38652964 DOI: 10.1016/j.intimp.2024.112129] [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: 10/30/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Lung injury in sepsis is caused by an excessive inflammatory response caused by the entry of pathogenic microorganisms into the body. It is also accompanied by the production of large amounts of ROS. Ferroptosis and mitochondrial dysfunction have also been shown to be related to sepsis. Finding suitable sepsis therapeutic targets is crucial for sepsis research. BTB domain-containing protein 7 (KBTBD7) is involved in regulating inflammatory responses, but its role and mechanism in the treatment of septic lung injury are still unclear. In this study, we evaluated the role and related mechanisms of KBTBD7 in septic lung injury. In in vitro studies, we established an in vitro model by inducing human alveolar epithelial cells with lipopolysaccharide (LPS) and found that KBTBD7 was highly expressed in the in vitro model. KBTBD7 knockdown could reduce the inflammatory response by inhibiting the secretion of pro-inflammatory factors and inhibit the production of ROS, ferroptosis and mitochondrial dysfunction. Mechanistic studies show that KBTBD7 interacts with FOXA1, promotes FOXA1 expression, and indirectly inhibits SLC7A11 transcription. In vivo studies have shown that knocking down KBTBD7 improves lung tissue damage in septic lung injury mice, inhibits inflammatory factors, ROS production and ferroptosis. Taken together, knockdown of KBTBD7 shows an alleviating effect on septic lung injury in vitro and in vivo, providing a potential therapeutic target for the treatment of septic lung injury.
Collapse
Affiliation(s)
- Xiang Li
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Zhao Lin
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - ShiYu Xu
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Ning Zhang
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Jun Zhou
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Bo Liao
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China.
| |
Collapse
|
11
|
Zheng Y, An H, Qi J, Li J. Recent progress in thiocarbazone metal complexes for cancer therapy via mitochondrial signalling pathway. Front Chem 2024; 12:1424022. [PMID: 38873408 PMCID: PMC11169589 DOI: 10.3389/fchem.2024.1424022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
Mitochondria are the energy factories of cells and are important targets for the development of novel tumour treatment strategies owing to their involvement in processes such as apoptosis, oxidative stress, and metabolic programming. Thiosemicarbazone metal complexes target mitochondria and reduce mitochondrial membrane potential. The breakdown of mitochondrial membrane potential is a key event in the early stage of apoptosis, which releases cytochrome C and other pro-apoptotic factors, activates the intracellular apoptotic enzyme cascade, and eventually causes irreversible apoptosis of tumour cells. Thiosemicarbazone metal complexes targeting the mitochondria have recently emerged as potential antitumour agents; therefore, this review describes the structural diversity of thiosemicarbazone metal [Fe(III), Cu(II), Ni(II), Zn(II), Ga(III), Pb(II), Au(III), and Ir(III)] complexes and explores their anti-tumour mechanisms that target mitochondrial pathways.
Collapse
Affiliation(s)
- Yunyun Zheng
- Medical School of Pingdingshan University, Pingdingshan, China
| | - Hangyi An
- Guangxi Key Laboratory of Green Chemical Materials and Safety Technology, College of Petroleum and Chemical Engineering, Beibu Gulf University, Qinzhou, China
| | - Jinxu Qi
- Medical School of Pingdingshan University, Pingdingshan, China
| | - Jiaming Li
- Guangxi Key Laboratory of Green Chemical Materials and Safety Technology, College of Petroleum and Chemical Engineering, Beibu Gulf University, Qinzhou, China
| |
Collapse
|
12
|
Chaurembo AI, Xing N, Chanda F, Li Y, Zhang HJ, Fu LD, Huang JY, Xu YJ, Deng WH, Cui HD, Tong XY, Shu C, Lin HB, Lin KX. Mitofilin in cardiovascular diseases: Insights into the pathogenesis and potential pharmacological interventions. Pharmacol Res 2024; 203:107164. [PMID: 38569981 DOI: 10.1016/j.phrs.2024.107164] [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: 11/30/2023] [Revised: 03/09/2024] [Accepted: 03/29/2024] [Indexed: 04/05/2024]
Abstract
The impact of mitochondrial dysfunction on the pathogenesis of cardiovascular disease is increasing. However, the precise underlying mechanism remains unclear. Mitochondria produce cellular energy through oxidative phosphorylation while regulating calcium homeostasis, cellular respiration, and the production of biosynthetic chemicals. Nevertheless, problems related to cardiac energy metabolism, defective mitochondrial proteins, mitophagy, and structural changes in mitochondrial membranes can cause cardiovascular diseases via mitochondrial dysfunction. Mitofilin is a critical inner mitochondrial membrane protein that maintains cristae structure and facilitates protein transport while linking the inner mitochondrial membrane, outer mitochondrial membrane, and mitochondrial DNA transcription. Researchers believe that mitofilin may be a therapeutic target for treating cardiovascular diseases, particularly cardiac mitochondrial dysfunctions. In this review, we highlight current findings regarding the role of mitofilin in the pathogenesis of cardiovascular diseases and potential therapeutic compounds targeting mitofilin.
Collapse
Affiliation(s)
- Abdallah Iddy Chaurembo
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia, Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Na Xing
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China.
| | - Francis Chanda
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia, Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Li
- Department of Cardiology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine (Zhongshan Hospital of Traditional Chinese Medicine), Zhongshan, Guangdong, China; Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hui-Juan Zhang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Li-Dan Fu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jian-Yuan Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yun-Jing Xu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia, Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Wen-Hui Deng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hao-Dong Cui
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Guizhou Medical University, Guiyang, Guizhou, China
| | - Xin-Yue Tong
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia, Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Chi Shu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Food Science College, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Han-Bin Lin
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia, Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Kai-Xuan Lin
- Department of Cardiology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine (Zhongshan Hospital of Traditional Chinese Medicine), Zhongshan, Guangdong, China; Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
13
|
Zhang B, Li Y, Liu N, Liu B. AP39, a novel mitochondria-targeted hydrogen sulfide donor ameliorates doxorubicin-induced cardiotoxicity by regulating the AMPK/UCP2 pathway. PLoS One 2024; 19:e0300261. [PMID: 38568919 PMCID: PMC10990198 DOI: 10.1371/journal.pone.0300261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/25/2024] [Indexed: 04/05/2024] Open
Abstract
Doxorubicin (DOX) is a broad-spectrum, highly effective antitumor agent; however, its cardiotoxicity has greatly limited its use. Hydrogen sulfide (H2S) is an endogenous gaseous transmitter that exerts cardioprotective effects via the regulation of oxidative stress and apoptosis and maintenance of mitochondrial function, among other mechanisms. AP39 is a novel mitochondria-targeted H2S donor that, at appropriate concentrations, attenuates intracellular oxidative stress damage, maintains mitochondrial function, and ameliorates cardiomyocyte injury. In this study, DOX-induced cardiotoxicity models were established using H9c2 cells and Sprague-Dawley rats to evaluate the protective effect of AP39 and its mechanisms of action. Both in vivo and in vitro experiments showed that DOX induces oxidative stress injury, apoptosis, and mitochondrial damage in cardiomyocytes and decreases the expression of p-AMPK/AMPK and UCP2. All DOX-induced changes were attenuated by AP39 treatment. Furthermore, the protective effect of AP39 was significantly attenuated by the inhibition of AMPK and UCP2. The results suggest that AP39 ameliorates DOX-induced cardiotoxicity by regulating the expression of AMPK/UCP2.
Collapse
Affiliation(s)
- Bin Zhang
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| | - Yangxue Li
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| | - Ning Liu
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| | - Bin Liu
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| |
Collapse
|
14
|
Behrendt I, Röder I, Will F, Michel G, Friedrich E, Grote D, Martin Z, Dötzer HP, Fasshauer M, Speckmann M, Kuntz S. Grape/Blueberry Anthocyanins and Their Gut-Derived Metabolites Attenuate LPS/Nigericin-Induced Inflammasome Activation by Inhibiting ASC Speck Formation in THP-1 Monocytes. Metabolites 2024; 14:203. [PMID: 38668331 PMCID: PMC11051782 DOI: 10.3390/metabo14040203] [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: 02/29/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammasomes are multi-protein complexes, which are formed in response to tissue injury, infections, and metabolic stress. However, aberrant inflammasome activation has been linked to several inflammatory diseases. Anthocyanins have been reported to attenuate NLR family pyrin domain-containing 3 (NLRP3) inflammasome activation, but the influence of grape/blueberry anthocyanins and especially their gut-derived metabolites on NLRP3 inflammasome activation in human monocytes remains unclear. Therefore, human leukemic monocytes (THP-1 cells, Tohoku Hospital Pediatrics-1 cells) were preincubated with different concentrations of grape/blueberry anthocyanins, homovanillyl alcohol, or 2,4,6-trihydroxybenzaldehyde (THBA) before the NLRP3 inflammasome was activated by lipopolysaccharide and/or nigericin. Apoptosis-associated speck-like protein containing a CARD (ASC) speck formation, as well as ASC and NLRP3 protein expression, were determined using flow cytometry. Caspase-1 activity was measured in cultured cells, and pro-inflammatory cytokine secretion was determined using enzyme-linked immunosorbent assays. Anthocyanins and their metabolites had no effect on ASC or NLRP3 protein expression. However, THBA significantly inhibited ASC speck formation in primed and unprimed THP-1 monocytes, while caspase-1 activity was significantly declined by grape/blueberry anthocyanins. Furthermore, reduced inflammasome activation resulted in lower pro-inflammatory cytokine secretion. In conclusion, our results show for the first time that grape/blueberry anthocyanins and their gut-derived metabolites exert anti-inflammatory effects by attenuating NLRP3 inflammasome activation in THP-1 monocytes.
Collapse
Affiliation(s)
- Inken Behrendt
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Isabella Röder
- Department of Beverage Research, Hochschule Geisenheim University, 65366 Geisenheim, Germany; (I.R.); (F.W.)
| | - Frank Will
- Department of Beverage Research, Hochschule Geisenheim University, 65366 Geisenheim, Germany; (I.R.); (F.W.)
| | - Gabriela Michel
- Institute for Clinical Immunology, Transfusion Medicine and Hemostaseology, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (G.M.); (M.S.)
- Flow Cytometry Core Facility, Department of Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Elvira Friedrich
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Daniela Grote
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Zoe Martin
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Hanna Pauline Dötzer
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Mathias Fasshauer
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Martin Speckmann
- Institute for Clinical Immunology, Transfusion Medicine and Hemostaseology, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (G.M.); (M.S.)
- Flow Cytometry Core Facility, Department of Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Sabine Kuntz
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| |
Collapse
|
15
|
Wang G, Ma F, Zhang W, Xin Y, Ping K, Wang Y, Dong J. Malvidin alleviates LPS-induced septic intestinal injury through the nuclear factor erythroid 2-related factor 2/reactive oxygen species/NLRP3 inflammasome pathway. Inflammopharmacology 2024; 32:893-901. [PMID: 38100033 DOI: 10.1007/s10787-023-01378-8] [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: 07/06/2023] [Accepted: 09/11/2023] [Indexed: 03/03/2024]
Abstract
Emerging evidence suggests that the gastrointestinal tract plays a crucial role in the pathophysiology of sepsis, a leading cause of mortality among patients admitted to the intensive care unit (ICU). Malvidin, belonging to the flavonoid family of compounds, exhibits a range of capabilities including anti-inflammatory and antioxidant properties. Studies have demonstrated that Malvidin exhibits a dose-dependent effect in mitigating sepsis-induced intestinal injury. The advantageous impact of Malvidin in safeguarding against sepsis-induced intestinal injury is associated with its capacity to counteract oxidative stress, inhibit cellular apoptosis, diminish the secretion of pro-inflammatory cytokines, and regulate the synthesis of inflammasomes. The findings indicate that Malvidin, a natural compound, exhibits protective effects on the gut by activating the nuclear factor erythroid 2-related factor 2/reactive oxygen species/NLRP3 inflammasome pathway. These results have significant implications for potential clinical applications and offer valuable insights into the treatment of sepsis-induced intestinal injury.
Collapse
Affiliation(s)
- Guanglu Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Fenfen Ma
- Department of Medicine Laboratory, Department of Cardiology, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang, China
| | - Wei Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yue Xin
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Kaixin Ping
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yan Wang
- Department of Medicine Laboratory, Department of Cardiology, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang, China.
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
16
|
Hao L, Li S, Chen G, Hu X. Regulation of UCP2 in nonalcoholic fatty liver disease: From mechanisms to natural product. Chem Biol Drug Des 2024; 103. [DOI: 10.1111/cbdd.14461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/09/2024] [Indexed: 01/04/2025]
Abstract
AbstractNonalcoholic fatty liver disease (NAFLD) is a chronic liver disease associated with lipid deposition in liver cells and/or subsequent inflammation, excluding other known causes. NAFLD is a subset of metabolic syndrome that ranges from simple steatohepatitis (NASH), fibrosis to cirrhosis and hepatocellular carcinoma (HCC). At present, the pathogenesis of NAFLD remains unclear. Among the many factors that shape these transitions, uncoupling protein 2 (UCP2) may be involved in every stage of the disease. UCP2 is a carrier protein that responds to fatty acids (FAs) in mitochondrial intima and has a wide tissue distribution. However, the biological function of UCP2 has not been fully elucidated, and most of our current knowledge comes from cell and animal experiments. These data suggest that UCP2 plays a role in lipid metabolism, oxidative stress, apoptosis, and even cancer. In this review, we summarize the structure, distribution, and biological function of UCP2 and its role in the progression of NAFLD, as well as natural products targeting UCP2 to improve NAFLD.
Collapse
Affiliation(s)
- Liyuan Hao
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
- Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Shenghao Li
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
- Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Guo Chen
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Xiaoyu Hu
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| |
Collapse
|
17
|
Strugała-Danak P, Spiegel M, Gabrielska J. Malvidin and Its Mono- and Di-Glucosides Forms: A Study of Combining Both In Vitro and Molecular Docking Studies Focused on Cholinesterase, Butyrylcholinesterase, COX-1 and COX-2 Activities. Molecules 2023; 28:7872. [PMID: 38067599 PMCID: PMC10708353 DOI: 10.3390/molecules28237872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Malvidin, one of the six most prominent anthocyanins found in various fruits and vegetables, may possess a wide range of health-promoting properties. The biological activity of malvidin and its glycosides is not entirely clear and has been relatively less frequently studied compared to other anthocyanins. Therefore, this study aimed to determine the relationship between the structural derivatives of malvidin and their anti-cholinergic and anti-inflammatory activity. The study selected malvidin (Mv) and its two sugar derivatives: malvidin 3-O-glucoside (Mv 3-glc) and malvidin 3,5-O-diglucoside (Mv 3,5-diglc). The anti-inflammatory activity was assessed by inhibiting the enzymes, specifically COX-1 and COX-2. Additionally, the inhibitory effects on cholinesterase activity, particularly acetylcholinesterase (AChE) and butyrylcholinesterase (BChE), were evaluated. Molecular modeling was also employed to examine and visualize the interactions between enzymes and anthocyanins. The results revealed that the highest inhibitory capacity at concentration 100 µM was demonstrated by Mv 3-glc in relation to AChE (26.3 ± 3.1%) and BChE (22.1 ± 3.0%), highlighting the crucial role of the glycoside substituent at the C3 position of the C ring in determining the inhibitory efficiency of these enzymes. In addition, the glycosylation of malvidin significantly reduced the anti-inflammatory activity of these derivatives compared to the aglycone form. The IC50 parameter demonstrates the following relationship for the COX-1 enzyme: Mv (12.45 ± 0.70 µM) < Mv 3-glc (74.78 ± 0.06 µM) < Mv 3,5-diglc (90.36 ± 1.92 µM). Similarly, for the COX-2 enzyme, we have: Mv (2.76 ± 0.16 µM) < Mv 3-glc (39.92 ± 3.02 µM) < Mv 3.5-diglc (66.45 ± 1.93 µM). All tested forms of malvidin exhibited higher activity towards COX-2 compared to COX-1, indicating their selectivity as inhibitors of COX-2. Theoretical calculations were capable of qualitatively replicating most of the noted patterns in the experimental data, explaining the impact of deprotonation and glycosylation on inhibitory activity. It can be suggested that anthocyanins, such as malvidins, could be valuable in the development of treatments for inflammatory conditions and Alzheimer's disease and deserve further study.
Collapse
Affiliation(s)
- Paulina Strugała-Danak
- Department of Physics and Biophysics, Wrocław University of Environmental and Life Sciences, C. K. Norwida 25, 50-375 Wrocław, Poland;
| | - Maciej Spiegel
- Department of Organic Chemistry and Pharmaceutical Technology, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland
| | - Janina Gabrielska
- Department of Physics and Biophysics, Wrocław University of Environmental and Life Sciences, C. K. Norwida 25, 50-375 Wrocław, Poland;
| |
Collapse
|
18
|
Hu J, Xie S, Zhang H, Wang X, Meng B, Zhang L. Microglial Activation: Key Players in Sepsis-Associated Encephalopathy. Brain Sci 2023; 13:1453. [PMID: 37891821 PMCID: PMC10605398 DOI: 10.3390/brainsci13101453] [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: 08/20/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a common brain dysfunction, which results in severe cognitive and neurological sequelae and an increased mortality rate in patients with sepsis. Depending on the stimulus, microglia (resident macrophages in the brain that are involved in SAE pathology and physiology) can adopt two polarization states (M1/M2), corresponding to altered microglial morphology, gene expression, and function. We systematically described the pathogenesis, morphology, function, and phenotype of microglial activation in SAE and demonstrated that microglia are closely related to SAE occurrence and development, and concomitant cognitive impairment. Finally, some potential therapeutic approaches that can prime microglia and neuroinflammation toward the beneficial restorative microglial phenotype in SAE were outlined.
Collapse
Affiliation(s)
- Jiyun Hu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shucai Xie
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Haisong Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xinrun Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Binbin Meng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care 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
|
19
|
Merecz-Sadowska A, Sitarek P, Kowalczyk T, Zajdel K, Jęcek M, Nowak P, Zajdel R. Food Anthocyanins: Malvidin and Its Glycosides as Promising Antioxidant and Anti-Inflammatory Agents with Potential Health Benefits. Nutrients 2023; 15:3016. [PMID: 37447342 DOI: 10.3390/nu15133016] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/23/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Anthocyanins are flavonoid compounds that are abundantly present in fruits and vegetables. These compounds contribute to the color of these foods and offer various health benefits to consumers due to their biological properties. There are more than 1000 types of anthocyanins in nature, all derived from 27 anthocyanidin aglycones that have different glycosylations and acylations. Malvidin is one of the most well-known anthocyanidins. Several studies, including those conducted on cell lines, animals, and humans, have suggested that malvidin and its glycosides possess anti-carcinogenic, diabetes-control, cardiovascular-disease-prevention, and brain-function-improvement properties. These health benefits are primarily attributed to their antioxidant and anti-inflammatory effects, which are influenced by the molecular mechanisms related to the expression and modulation of critical genes. In this article, we review the available information on the biological activity of malvidin and its glycosides concerning their health-promoting effects.
Collapse
Affiliation(s)
- Anna Merecz-Sadowska
- Department of Economic and Medical Informatics, University of Lodz, 90-214 Lodz, Poland
| | - Przemysław Sitarek
- Department of Medical Biology, Medical University of Lodz, 90-151 Lodz, Poland
| | - Tomasz Kowalczyk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
| | - Karolina Zajdel
- Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland
| | - Mariusz Jęcek
- Department of Economic and Medical Informatics, University of Lodz, 90-214 Lodz, Poland
| | - Paweł Nowak
- Department of Economic and Medical Informatics, University of Lodz, 90-214 Lodz, Poland
| | - Radosław Zajdel
- Department of Economic and Medical Informatics, University of Lodz, 90-214 Lodz, Poland
| |
Collapse
|