1
|
Li Z, Shang D. NOD1 and NOD2: Essential Monitoring Partners in the Innate Immune System. Curr Issues Mol Biol 2024; 46:9463-9479. [PMID: 39329913 PMCID: PMC11430502 DOI: 10.3390/cimb46090561] [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: 05/29/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Nucleotide-binding oligomerization domain containing 1 (NOD1) and NOD2 are pivotal cytoplasmic pattern-recognition receptors (PRRs) that exhibit remarkable evolutionary conservation. They possess the ability to discern specific peptidoglycan (PGN) motifs, thereby orchestrating innate immunity and contributing significantly to immune homeostasis maintenance. The comprehensive understanding of both the structure and function of NOD1 and NOD2 has been extensively elucidated. These receptors proficiently recognize an array of damage-associated molecular patterns (DAMPs) as well as pathogen-associated molecular patterns (PAMPs), subsequently mediating inflammatory responses and autophagy. In recent years, emerging evidence has highlighted the crucial roles played by NOD1 and NOD2 in regulating infectious diseases, metabolic disorders, cancer, and autoimmune conditions, among others. Perturbation in either their loss or excessive activation can detrimentally impact immune homeostasis. This review offers a comprehensive overview of the structural characteristics, subcellular localization, activation mechanisms, and significant roles of NOD1 and NOD2 in innate immunity and related disease.
Collapse
Affiliation(s)
- Zhenjia Li
- School of Life Science, Liaoning Normal University, Dalian 116081, China
| | - Dejing Shang
- School of Life Science, Liaoning Normal University, Dalian 116081, China
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian 116081, China
| |
Collapse
|
2
|
Su H, Wang X, Wang L, Yuan N. Therapeutic Targeting of Pattern Recognition Receptors to Modulate Inflammation in Atherosclerosis. Cell Biochem Biophys 2024:10.1007/s12013-024-01481-9. [PMID: 39145823 DOI: 10.1007/s12013-024-01481-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
Atherosclerosis (AS), a potentially fatal cardiovascular disease (CVD), is a chronic inflammatory condition. The disease's onset and progression are influenced by inflammatory and immunological mechanisms. The innate immune pathways are essential in the progression of AS, as they are responsible for detecting first danger signals and causing long-term changes in immune cells. The innate immune system possesses distinct receptors known as pattern recognition receptors (PRRs) which can identify both pathogen-associated molecular patterns and danger-associated molecular signals. Activation of PRRs initiates the inflammatory response in various physiological systems, such as the cardiovascular system. This review specifically examines the contribution of the innate immune response and PRRs to the formation and advancement of AS. Studying the role of these particular receptors in AS would enhance our understanding of the development of AS and offer novel approaches for directly improving the inflammatory response associated with it.
Collapse
Affiliation(s)
- Hongyan Su
- Cardiology Department, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130000, China
| | - Xiancheng Wang
- Cardiology Department, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130000, China
| | - Lu Wang
- Cardiology Department, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130000, China
| | - Na Yuan
- Rheumatology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 30000, China.
| |
Collapse
|
3
|
Fan J, Zhu T, Tian X, Liu S, Zhang SL. Exploration of ferroptosis and necroptosis-related genes and potential molecular mechanisms in psoriasis and atherosclerosis. Front Immunol 2024; 15:1372303. [PMID: 39072329 PMCID: PMC11272566 DOI: 10.3389/fimmu.2024.1372303] [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: 01/17/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024] Open
Abstract
Objective Ferroptosis and necroptosis are two recently identified forms of non-apoptotic cell death. Their dysregulation plays a critical role in the development and progression of Psoriasis (PsD) and Atherosclerosis (AS). This study explores shared Ferroptosis and necroptosis-related genes and elucidates their molecular mechanisms in PsD and AS through the analysis of public databases. Methods Data sets for PsD (GSE30999) and AS (GSE28829) were retrieved from the GEO database. Differential gene expression (DEG) and weighted gene co-expression network analysis (WGCNA) were performed. Machine learning algorithms identified candidate biomarkers, whose diagnostic values were assessed using Receiver Operating Characteristic (ROC) curve analysis. Additionally, the expression levels of these biomarkers in cell models of AS and PsD were quantitatively measured using Western Blot (WB) and real-time quantitative PCR (RT-qPCR). Furthermore, CIBERSORT evaluated immune cell infiltration in PsD and AS tissues, highlighting the correlation between characteristic genes and immune cells. Predictive analysis for candidate drugs targeting characteristic genes was conducted using the DGIdb database, and an lncRNA-miRNA-mRNA network related to these genes was constructed. Results We identified 44 differentially expressed ferroptosis-related genes (DE-FRGs) and 30 differentially expressed necroptosis-related genes (DE-NRGs). GO and KEGG enrichment analyses revealed significant enrichment of these genes in immune-related and inflammatory pathways, especially in NOD-like receptor and TNF signaling pathways. Two ferroptosis-related genes (NAMPT, ZFP36) and eight necroptosis-related genes (C7, CARD6, CASP1, CTSD, HMOX1, NOD2, PYCARD, TNFRSF21) showed high sensitivity and specificity in ROC curve analysis. These findings were corroborated in external validation datasets and cell models. Immune infiltration analysis revealed increased levels of T cells gamma delta, Macrophages M0, and Macrophages M2 in PsD and AS samples. Additionally, we identified 43 drugs targeting 5 characteristic genes. Notably, the XIST-miR-93-5p-ZFP36/HMOX1 and NEAT1-miR-93-5p-ZFP36/HMOX1 pathways have been identified as promising RNA regulatory pathways in AS and PsD. Conclusion The two ferroptosis-related genes (NAMPT, ZFP36) and eight necroptosis-related genes (C7, CARD6, CASP1, CTSD, HMOX1, NOD2, PYCARD, TNFRSF21) are potential key biomarkers for PsD and AS. These genes significantly influence the pathogenesis of PsD and AS by modulating macrophage activity, participating in immune regulation, and mediating inflammatory responses.
Collapse
Affiliation(s)
- Jilin Fan
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tingting Zhu
- Department of Neurosurgery Ward 5, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiaoling Tian
- Department of Neurosurgery Ward 5, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Sijia Liu
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Shi-Liang Zhang
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
4
|
Haybar H, Sarbazjoda E, Purrahman D, Mahmoudian-Sani MR, Saki N. The prognostic potential of long noncoding RNA XIST in cardiovascular diseases: a review. Per Med 2024; 21:257-269. [PMID: 38889283 DOI: 10.1080/17410541.2024.2360380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
There is a significant mortality rate associated with cardiovascular disease despite advances in treatment. long Non-coding RNAs (lncRNAs) play a critical role in many biological processes and their dysregulation is associated with a wide range of diseases in which their downstream pathways are disrupted. A lncRNA X-inactive specific transcript (XIST) is well known as a factor that regulates the physiological process of chromosome dosage compensation for females. According to recent studies, lncRNA XIST is involved in a variety of cellular processes, including apoptosis, proliferation, invasion, metastasis, oxidative stress and inflammation, through molecular networks with microRNAs and their downstream targets in neoplastic and non-neoplastic diseases. Because these cellular processes play a role in the pathogenesis of cardiovascular diseases, we aim to investigate the role that lncRNA XIST plays in this process. Additionally, we wish to determine whether it is a prognostic factor or a potential therapeutic target in these diseases.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ehsan Sarbazjoda
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz,Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Daryush Purrahman
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz,Iran
| | - Mohammad Reza Mahmoudian-Sani
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz,Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz,Iran
| |
Collapse
|
5
|
Kong L, Cao Y, He Y, Zhang Y. Role and molecular mechanism of NOD2 in chronic non-communicable diseases. J Mol Med (Berl) 2024; 102:787-799. [PMID: 38740600 DOI: 10.1007/s00109-024-02451-7] [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: 09/11/2023] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024]
Abstract
Nucleotide-binding oligomerization domain containing 2 (NOD2), located in the cell cytoplasm, is a pattern recognition receptor belonging to the innate immune receptor family. It mediates the innate immune response by identifying conserved sequences in bacterial peptide glycans and plays an essential role in maintaining immune system homeostasis. Gene mutations of NOD2 lead to the development of autoimmune diseases such as Crohn's disease and Blau syndrome. Recently, NOD2 has been shown to be associated with the pathogenesis of diabetes, cardiac-cerebral diseases, and cancers. However, the function of NOD2 in these non-communicable diseases (CNCDs) is not well summarized in reviews. Our report mainly discusses the primary function and molecular mechanism of NOD2 as well as its potential clinical significance in CNCDs.
Collapse
Affiliation(s)
- Lingjun Kong
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan, Shandong, People's Republic of China
| | - Yanhua Cao
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan, Shandong, People's Republic of China
| | - Yanan He
- Gamma Knife Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Yahui Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
6
|
Luo JM, Lin HB, Weng YQ, Lin YH, Lai LY, Li J, Li FX, Xu SY, Zhang HF, Zhao W. Inhibition of PARP1 improves cardiac function after myocardial infarction via up-regulated NLRC5. Chem Biol Interact 2024; 395:111010. [PMID: 38679114 DOI: 10.1016/j.cbi.2024.111010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/30/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024]
Abstract
The incidence and mortality rate of myocardial infarction are increasing per year in China. The polarization of macrophages towards the classically activated macrophages (M1) phenotype is of utmost importance in the progression of inflammatory stress subsequent to myocardial infarction. Poly (ADP-ribose) polymerase 1(PARP1) is the ubiquitous and best characterized member of the PARP family, which has been reported to support macrophage polarization towards the pro-inflammatory phenotype. Yet, the role of PARP1 in myocardial ischemic injury remains to be elucidated. Here, we demonstrated that a myocardial infarction mouse model induced cardiac damage characterized by cardiac dysfunction and increased PARP1 expression in cardiac macrophages. Inhibition of PARP1 by the PJ34 inhibitors could effectively alleviate M1 macrophage polarization, reduce infarction size, decrease inflammation and rescue the cardiac function post-MI in mice. Mechanistically, the suppression of PARP1 increase NLRC5 gene expression, and thus inhibits the NF-κB pathway, thereby decreasing the production of inflammatory cytokines such as IL-1β and TNF-α. Inhibition of NLRC5 promote infection by effectively abolishing the influence of this mechanism discussed above. Interestingly, inhibition of NLRC5 promotes cardiac macrophage polarization toward an M1 phenotype but without having major effects on M2 macrophages. Our results demonstrate that inhibition of PARP1 increased NLRC5 gene expression, thereby suppressing M1 polarization, improving cardiac function, decreasing infarct area and attenuating inflammatory injury. The aforementioned findings provide new insights into the proinflammatory mechanisms that drive macrophage polarization following myocardial infarction, thereby introducing novel potential targets for future therapeutic interventions in individuals affected by myocardial infarction.
Collapse
Affiliation(s)
- Jia-Ming Luo
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, China
| | - Hong-Bin Lin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, China
| | - Ya-Qian Weng
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, China
| | - Ying-Hui Lin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, China; Department of Anesthesiology, The First Affiliated Hospital of Shantou University Medical College, Guangdong Province, China
| | - Lu-Ying Lai
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, China
| | - Ji Li
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, China
| | - Feng-Xian Li
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, China
| | - Shi-Yuan Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, China
| | - Hong-Fei Zhang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, China.
| | - Wei Zhao
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, China.
| |
Collapse
|
7
|
Qin H, Yang F, Hao P, Zhang X. Gut microbiota and cerebrovascular diseases: a Mendelian randomization study. Front Microbiol 2023; 14:1228815. [PMID: 37637136 PMCID: PMC10448814 DOI: 10.3389/fmicb.2023.1228815] [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: 05/25/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Background The causal relationship between gut microbiota and cerebrovascular disease remains unknown, despite several recent studies reporting an association between the two. Methods To assess this relationship, we conducted a two-sample Mendelian randomization (MR) using summary statistics data from published genome-wide association studies (GWAS). This analysis allowed us to identify bacterial taxa that may affect cerebrovascular disease. Furthermore, we performed reverse MR to further analyze the significant bacterial taxa. Finally, we conducted a two-step MR analysis to examine the mediating role of metabolic factors [systolic blood pressure (SBP), type 2 diabetes (T2D), and body mass index (BMI)] in the association between gut microbiota and cerebrovascular disease. Additionally, a series of sensitivity analyses were carried out to validate the robustness of our findings. Results Our results showed that a genetically predicted high abundance of family Porphyromonadaceae reduced the risk of intracranial aneurysms (IA). Moreover, using inverse variance weighted (IVW) estimates, we found a nominal causal relationship between seventeen gut microbiota and IA, as well as its subtypes. In the case of stroke and its subtypes, we observed a nominal causal relationship with thirteen, eleven, eleven, nine, and eight bacteria for AS, AIS, CES, LAS, and SVS, respectively. Reverse MR analysis showed no significant causal relationship between intracranial aneurysms and gut microbiota. However, we did find that genetically predicted any stroke (AS) and any ischemic stroke (AIS) reduced the abundance of family Clostridiaceae1 (OR: 0.74, 95% CI: 0.62-0.87, p = 3.39 × 10-4, and OR: 0.75, 95% CI: 0.66-0.87, p = 7.06 × 10-5, respectively). Furthermore, genetic prediction of AIS (OR: 0.87, 95% CI: 0.77-0.99, p = 3.05 × 10-2) was associated with a reduced abundance of the order Clostridiales. Moreover, genus Streptococcus exhibited effects on AS, AIS, and SVS which were mediated by T2D. Conversely, the association between genus Eubacterium brachy group and AIS was mediated by SBP. No significant heterogeneity of instrumental variables or horizontal pleiotropy was observed. Conclusion This MR analysis indicates that there exists a beneficial or detrimental causal effect of gut microbiota composition on cerebrovascular disease. And SBP and T2D may play mediating role in this process.
Collapse
Affiliation(s)
- Hao Qin
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Fan Yang
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Pengfei Hao
- Department of Neurosurgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xianfeng Zhang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Yu F, Duan Y, Liu C, Huang H, Xiao X, He Z. Extracellular vesicles in atherosclerosis and vascular calcification: the versatile non-coding RNAs from endothelial cells and vascular smooth muscle cells. Front Med (Lausanne) 2023; 10:1193660. [PMID: 37469665 PMCID: PMC10352799 DOI: 10.3389/fmed.2023.1193660] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/12/2023] [Indexed: 07/21/2023] Open
Abstract
Atherosclerosis (AS) is characterized by the accumulation of lipids, fibrous elements, and calcification in the innermost layers of arteries. Vascular calcification (VC), the deposition of calcium and phosphate within the arterial wall, is an important characteristic of AS natural history. However, medial arterial calcification (MAC) differs from intimal calcification and cannot simply be explained as the consequence of AS. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are directly involved in AS and VC processes. Understanding the communication between ECs and VSMCs is critical in revealing mechanisms underlying AS and VC. Extracellular vesicles (EVs) are found as intercellular messengers in kinds of physiological processes and pathological progression. Non-coding RNAs (ncRNAs) encapsulated in EVs are involved in AS and VC, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). The effects of ncRNAs have not been comprehensively understood, especially encapsulated in EVs. Some ncRNAs have demonstrated significant roles in AS and VC, but it remains unclear the functions of the majority ncRNAs detected in EVs. In this review, we summarize ncRNAs encapsulated in EC-EVs and VSMC-EVs, and the signaling pathways that are involved in AS and VC.
Collapse
Affiliation(s)
- Fengyi Yu
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yingjie Duan
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chongmei Liu
- Department of Pathology, Yueyang People's Hospital, Yueyang, Hunan, China
| | - Hong Huang
- Hengyang Medical School, The First Affiliated Hospital, Institute of Clinical Medicine, University of South China, Hengyang, Hunan, China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhangxiu He
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
9
|
Wang Y, Zheng Y, Liu Y, Shan G, Zhang B, Cai Q, Lou J, Qu Y. The lipid-lowering effects of fenugreek gum, hawthorn pectin, and burdock inulin. Front Nutr 2023; 10:1149094. [PMID: 37032784 PMCID: PMC10076561 DOI: 10.3389/fnut.2023.1149094] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Objective The present study aimed to investigate the lipid-lowering effects and mechanisms of fenugreek gum (FG), hawthorn pectin (HP), and burdock inulin (BI) on high-fat diet (HFD)-induced hyperlipidemic rats. Methods In this study, high-fat diet (HFD) together with fat emulsion administration were used to establish hyperlipidemia model. The biochemical indices were assayed after administration of FG, HP, and BI. Their effects were evaluated by factor analysis. Alterations of gut microbiota and short chain fatty acids (SCFAs) in the cecal were assessed to illustrate the mechanism of lipid lowering. Results The supplementation of FG, HP, and BI on HFD-fed rats decreased the levels of serum lipid and reduced the HFD-related liver and testicle damage. In the scatter plot of factor analysis, HP and BI were closer to normal fat diet (NFD) group in restoring the severity of hyperlipidemia, while FG and HP enhanced the excretion of cholesterol and bile acids (BAs). The levels of total SCFAs, especially butyric acid reduced by HFD were increased by HP. The ratio of Firmicutes to Bacteroidetes increased by HFD was reduced by HP and BI. FG, HP, and BI enriched intestinal probiotics, which were related to bile acid excretion or lipid-lowering. Conclusions FG inhibited the absorption of cholesterol and enhanced the excretion of it, as well as increased the abundance of beneficial bacteria. While BI restored the imbalance of intestinal microbiota. HP enhanced the excretion of cholesterol and BAs, and restored the imbalance of intestinal microbiota. It was also utilized by intestinal microorganisms to yield SCFAs. This study suggested that FG, HP, and BI possessed the potential to be utilized as dietary supplements for obesity management.
Collapse
|
10
|
Wang L, Wang S, Zhang Q, He C, Fu C, Wei Q. The role of the gut microbiota in health and cardiovascular diseases. MOLECULAR BIOMEDICINE 2022; 3:30. [PMID: 36219347 PMCID: PMC9554112 DOI: 10.1186/s43556-022-00091-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
The gut microbiota is critical to human health, such as digesting nutrients, forming the intestinal epithelial barrier, regulating immune function, producing vitamins and hormones, and producing metabolites to interact with the host. Meanwhile, increasing evidence indicates that the gut microbiota has a strong correlation with the occurrence, progression and treatment of cardiovascular diseases (CVDs). In patients with CVDs and corresponding risk factors, the composition and ratio of gut microbiota have significant differences compared with their healthy counterparts. Therefore, gut microbiota dysbiosis, gut microbiota-generated metabolites, and the related signaling pathway may serve as explanations for some of the mechanisms about the occurrence and development of CVDs. Several studies have also demonstrated that many traditional and latest therapeutic treatments of CVDs are associated with the gut microbiota and its generated metabolites and related signaling pathways. Given that information, we summarized the latest advances in the current research regarding the effect of gut microbiota on health, the main cardiovascular risk factors, and CVDs, highlighted the roles and mechanisms of several metabolites, and introduced corresponding promising treatments for CVDs regarding the gut microbiota. Therefore, this review mainly focuses on exploring the role of gut microbiota related metabolites and their therapeutic potential in CVDs, which may eventually provide better solutions in the development of therapeutic treatment as well as the prevention of CVDs.
Collapse
Affiliation(s)
- Lu Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Shiqi Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Qing Zhang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chengqi He
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chenying Fu
- grid.412901.f0000 0004 1770 1022National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,grid.412901.f0000 0004 1770 1022Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Quan Wei
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| |
Collapse
|
11
|
Homeostasis in the Gut Microbiota in Chronic Kidney Disease. Toxins (Basel) 2022; 14:toxins14100648. [PMID: 36287917 PMCID: PMC9610479 DOI: 10.3390/toxins14100648] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
The gut microbiota consists of trillions of microorganisms, fulfilling important roles in metabolism, nutritional intake, physiology and maturation of the immune system, but also aiding and abetting the progression of chronic kidney disease (CKD). The human gut microbiome consists of bacterial species from five major bacterial phyla, namely Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia. Alterations in the members of these phyla alter the total gut microbiota, with a decline in the number of symbiotic flora and an increase in the pathogenic bacteria, causing or aggravating CKD. In addition, CKD-associated alteration of this intestinal microbiome results in metabolic changes and the accumulation of amines, indoles and phenols, among other uremic metabolites, which have a feedforward adverse effect on CKD patients, inhibiting renal functions and increasing comorbidities such as atherosclerosis and cardiovascular diseases (CVD). A classification of uremic toxins according to the degree of known toxicity based on the experimental evidence of their toxicity (number of systems affected) and overall experimental and clinical evidence was selected to identify the representative uremic toxins from small water-soluble compounds, protein-bound compounds and middle molecules and their relation to the gut microbiota was summarized. Gut-derived uremic metabolites accumulating in CKD patients further exhibit cell-damaging properties, damage the intestinal epithelial cell wall, increase gut permeability and lead to the translocation of bacteria and endotoxins from the gut into the circulatory system. Elevated levels of endotoxins lead to endotoxemia and inflammation, further accelerating CKD progression. In recent years, the role of the gut microbiome in CKD pathophysiology has emerged as an important aspect of corrective treatment; however, the mechanisms by which the gut microbiota contributes to CKD progression are still not completely understood. Therefore, this review summarizes the current state of research regarding CKD and the gut microbiota, alterations in the microbiome, uremic toxin production, and gut epithelial barrier degradation.
Collapse
|
12
|
Novel Scaffolds for Modulation of NOD2 Identified by Pharmacophore-Based Virtual Screening. Biomolecules 2022; 12:biom12081054. [PMID: 36008948 PMCID: PMC9405794 DOI: 10.3390/biom12081054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 12/04/2022] Open
Abstract
Nucleotide-binding oligomerization domain-containing protein 2 (NOD2) is an innate immune pattern recognition receptor responsible for the recognition of bacterial peptidoglycan fragments. Given its central role in the formation of innate and adaptive immune responses, NOD2 represents a valuable target for modulation with agonists and antagonists. A major challenge in the discovery of novel small-molecule NOD2 modulators is the lack of a co-crystallized complex with a ligand, which has limited previous progress to ligand-based design approaches and high-throughput screening campaigns. To that end, a hybrid docking and pharmacophore modeling approach was used to identify key interactions between NOD2 ligands and residues in the putative ligand-binding site. Following docking of previously reported NOD2 ligands to a homology model of human NOD2, a structure-based pharmacophore model was created and used to virtually screen a library of commercially available compounds. Two compounds, 1 and 3, identified as hits by the pharmacophore model, exhibited NOD2 antagonist activity and are the first small-molecule NOD2 modulators identified by virtual screening to date. The newly identified NOD2 antagonist scaffolds represent valuable starting points for further optimization.
Collapse
|
13
|
Hu W, Kong X, Wang H, Li Y, Luo Y. Ischemic stroke and intestinal flora: an insight into brain-gut axis. Eur J Med Res 2022; 27:73. [PMID: 35614480 PMCID: PMC9131669 DOI: 10.1186/s40001-022-00691-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/21/2022] [Indexed: 11/10/2022] Open
Abstract
Stroke is a type of cerebrovascular disease that significantly endangers human health and lowers quality of life. This understandably places a heavy burden on society and families. In recent years, intestinal flora has attracted increasing attention from scholars worldwide, and its association with ischemic stroke is becoming a hot topic of research amongst researchers in field of stroke. After suffering from a stroke, intestinal microbial dysbiosis leads to increased intestinal permeability and activation of the intestinal immune system, which in turn leads to ectopic intestinal bacteria and pro-inflammatory cells that enter brain tissue through the damaged blood-brain barrier. This exacerbates ischemia-reperfusion injury. Interestingly, after a stroke, some metabolites produced by the intestinal flora attenuate ischemia-reperfusion injury by suppressing the post-stroke inflammatory response and promotes the repair of neurological function. Here we elucidate the changes in gut flora after occurrence of a stroke and highlight the immunomodulatory processes of the post-stroke gut flora.
Collapse
Affiliation(s)
- Wenjie Hu
- Department of Biological Science, Jining Medical University, Rizhao, Shandong, China.,Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
| | - Xiangyi Kong
- Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
| | - Hui Wang
- Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
| | - Yunqing Li
- Department of Pathogenic Biology, Jining Medical University, Jining, Shandong, China
| | - Yimin Luo
- Department of Biological Science, Jining Medical University, Rizhao, Shandong, China.
| |
Collapse
|
14
|
Ma X, Wang S, Cheng H, Ouyang H, Ma X. Melatonin Attenuates Ischemia/Reperfusion-Induced Oxidative Stress by Activating Mitochondrial Fusion in Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7105181. [PMID: 35047108 PMCID: PMC8763517 DOI: 10.1155/2022/7105181] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/20/2021] [Accepted: 10/01/2021] [Indexed: 12/31/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury can stimulate mitochondrial reactive oxygen species production. Optic atrophy 1- (OPA1-) induced mitochondrial fusion is an endogenous antioxidative mechanism that preserves the mitochondrial function. In our study, we investigated whether melatonin augments OPA1-dependent mitochondrial fusion and thus maintains redox balance during myocardial I/R injury. In hypoxia/reoxygenation- (H/R-) treated H9C2 cardiomyocytes, melatonin treatment upregulated OPA1 mRNA and protein expression, thereby enhancing mitochondrial fusion. Melatonin also suppressed apoptosis in H/R-treated cardiomyocytes, as evidenced by increased cell viability, diminished caspase-3 activity, and reduced Troponin T secretion; however, silencing OPA1 abolished these effects. H/R treatment augmented mitochondrial ROS production and repressed antioxidative molecule levels, while melatonin reversed these changes in an OPA1-dependent manner. Melatonin also inhibited mitochondrial permeability transition pore opening and maintained the mitochondrial membrane potential, but OPA1 silencing prevented these outcomes. These results illustrate that melatonin administration alleviates cardiomyocyte I/R injury by activating OPA1-induced mitochondrial fusion and inhibiting mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Xiaoling Ma
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| | - Shengchi Wang
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| | - Hui Cheng
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| | - Haichun Ouyang
- Department of Cardiology, The Seventh Affiliated Hospital, Southern Medical University, China
| | - Xiaoning Ma
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
15
|
Chen J, Li F, Yang W, Jiang S, Li Y. Comparison of Gut Microbiota and Metabolic Status of Sows With Different Litter Sizes During Pregnancy. Front Vet Sci 2021; 8:793174. [PMID: 35004929 PMCID: PMC8733392 DOI: 10.3389/fvets.2021.793174] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/19/2021] [Indexed: 01/07/2023] Open
Abstract
The experiment was conducted to compare the differences of gut microbiota and metabolic status of sows with different litter sizes on days 30 and 110 of gestation, and uncover the relationship between the composition of maternal gut microbiota during gestation and sow reproductive performance. Twenty-six Large White × Landrace crossbred multiparous sows (2nd parity) with similar back fat thickness and body weight were assigned to two groups [high-reproductive performance group (HP group) and low-reproductive performance group (LP group)] according to their litter sizes and fed a common gestation diet. Results showed that compared with LP sows, HP sows had significantly lower plasma levels of triglyceride (TG) on gestation d 30 (P < 0.05), but had significantly higher plasma levels of TG, non-esterified fatty acid, tumor necrosis factor-α, and immunoglobulin M on gestation d 110 (P < 0.05). Consistently, HP sows revealed increased alpha diversity and butyrate-producing genera, as well as fecal butyrate concentration, on gestation d 30; HP sows showed significantly different microbiota community structure with LP sows (P < 0.05) and had markedly higher abundance of Firmicutes (genera Christensenellaceae_R-7_group and Terrisporobacter) which were positively related with litter size on gestation d 110 than LP sows (P < 0.05). In addition, plasma biochemical parameters, plasma cytokines, and fecal microbiota shifted dramatically from gestation d 30 to d 110. Therefore, our findings demonstrated that microbial abundances and community structures differed significantly between sows with different litter sizes and gestation stages, which was associated with changes in plasma biochemical parameters, inflammatory factors, and immunoglobulin. Moreover, these findings revealed that there was a significant correlation between litter size and gut microbiota of sows, and provided a microbial perspective to improve sow reproductive performance in pig production.
Collapse
Affiliation(s)
| | | | | | | | - Yang Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| |
Collapse
|
16
|
Fernández-García V, González-Ramos S, Martín-Sanz P, García-Del Portillo F, Laparra JM, Boscá L. NOD1 in the interplay between microbiota and gastrointestinal immune adaptations. Pharmacol Res 2021; 171:105775. [PMID: 34273489 DOI: 10.1016/j.phrs.2021.105775] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/30/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
Nucleotide-binding oligomerization domain 1 (NOD1), a pattern recognition receptor (PRR) that detects bacterial peptidoglycan fragments and other danger signals, has been linked to inflammatory pathologies. NOD1, which is expressed by immune and non-immune cells, is activated after recognizing microbe-associated molecular patterns (MAMPs). This recognition triggers host defense responses and both immune memory and tolerance can also be achieved during these processes. Since the gut microbiota is currently considered a master regulator of human physiology central in health and disease and the intestine metabolizes a wide range of nutrients, drugs and hormones, it is a fact that dysbiosis can alter tissues and organs homeostasis. These systemic alterations occur in response to gastrointestinal immune adaptations that are not yet fully understood. Even if previous evidence confirms the connection between the microbiota, the immune system and metabolic disorders, much remains to be discovered about the contribution of NOD1 to low-grade inflammatory pathologies such as obesity, diabetes and cardiovascular diseases. This review compiles the most recent findings in this area, while providing a dynamic and practical framework with future approaches for research and clinical applications on targeting NOD1. This knowledge can help to rate the consequences of the disease and to stratify the patients for therapeutic interventions.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | | | - José Moisés Laparra
- Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra, Cantoblanco 8, 28049 Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| |
Collapse
|
17
|
Li D, Wu M. Pattern recognition receptors in health and diseases. Signal Transduct Target Ther 2021; 6:291. [PMID: 34344870 PMCID: PMC8333067 DOI: 10.1038/s41392-021-00687-0] [Citation(s) in RCA: 641] [Impact Index Per Article: 213.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/23/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
Pattern recognition receptors (PRRs) are a class of receptors that can directly recognize the specific molecular structures on the surface of pathogens, apoptotic host cells, and damaged senescent cells. PRRs bridge nonspecific immunity and specific immunity. Through the recognition and binding of ligands, PRRs can produce nonspecific anti-infection, antitumor, and other immunoprotective effects. Most PRRs in the innate immune system of vertebrates can be classified into the following five types based on protein domain homology: Toll-like receptors (TLRs), nucleotide oligomerization domain (NOD)-like receptors (NLRs), retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), C-type lectin receptors (CLRs), and absent in melanoma-2 (AIM2)-like receptors (ALRs). PRRs are basically composed of ligand recognition domains, intermediate domains, and effector domains. PRRs recognize and bind their respective ligands and recruit adaptor molecules with the same structure through their effector domains, initiating downstream signaling pathways to exert effects. In recent years, the increased researches on the recognition and binding of PRRs and their ligands have greatly promoted the understanding of different PRRs signaling pathways and provided ideas for the treatment of immune-related diseases and even tumors. This review describes in detail the history, the structural characteristics, ligand recognition mechanism, the signaling pathway, the related disease, new drugs in clinical trials and clinical therapy of different types of PRRs, and discusses the significance of the research on pattern recognition mechanism for the treatment of PRR-related diseases.
Collapse
Affiliation(s)
- Danyang Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
18
|
Ji H, Wu D, Kimberlee O, Li R, Qian G. Molecular Perspectives of Mitophagy in Myocardial Stress: Pathophysiology and Therapeutic Targets. Front Physiol 2021; 12:700585. [PMID: 34276422 PMCID: PMC8279814 DOI: 10.3389/fphys.2021.700585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/27/2021] [Indexed: 01/15/2023] Open
Abstract
A variety of complex risk factors and pathological mechanisms contribute to myocardial stress, which ultimately promotes the development of cardiovascular diseases, including acute cardiac insufficiency, myocardial ischemia, myocardial infarction, high-glycemic myocardial injury, and acute alcoholic cardiotoxicity. Myocardial stress is characterized by abnormal metabolism, excessive reactive oxygen species production, an insufficient energy supply, endoplasmic reticulum stress, mitochondrial damage, and apoptosis. Mitochondria, the main organelles contributing to the energy supply of cardiomyocytes, are key determinants of cell survival and death. Mitophagy is important for cardiomyocyte function and metabolism because it removes damaged and aged mitochondria in a timely manner, thereby maintaining the proper number of normal mitochondria. In this review, we first introduce the general characteristics and regulatory mechanisms of mitophagy. We then describe the three classic mitophagy regulatory pathways and their involvement in myocardial stress. Finally, we discuss the two completely opposite effects of mitophagy on the fate of cardiomyocytes. Our summary of the molecular pathways underlying mitophagy in myocardial stress may provide therapeutic targets for myocardial protection interventions.
Collapse
Affiliation(s)
- Haizhe Ji
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China.,Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dan Wu
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| | - O'Maley Kimberlee
- School of Public Health, University of California, Berkeley, Berkeley, CA, United States
| | - Ruibing Li
- Department of Clinical Laboratory Medicine, The First Medical Center, Medical School of Chinese People's Liberation Army, Beijing, China
| | - Geng Qian
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
19
|
Protective Effect of Optic Atrophy 1 on Cardiomyocyte Oxidative Stress: Roles of Mitophagy, Mitochondrial Fission, and MAPK/ERK Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3726885. [PMID: 34211623 PMCID: PMC8205577 DOI: 10.1155/2021/3726885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/08/2021] [Accepted: 05/21/2021] [Indexed: 12/14/2022]
Abstract
Myocardial infarction is associated with oxidative stress and mitochondrial damage. However, the regulatory mechanisms underlying cardiomyocyte oxidative stress during myocardial infarction are not fully understood. In the present study, we explored the cardioprotective action of optic atrophy 1- (Opa1-) mediated mitochondrial autophagy (mitophagy) in oxidative stress-challenged cardiomyocytes, with a focus on mitochondrial homeostasis and the MAPK/ERK pathway. Our results demonstrated that overexpression of Opa1 in cultured rat H9C2 cardiomyocytes, a procedure that stimulates mitophagy, attenuates oxidative stress and increases cellular antioxidant capacity. Activation of Opa1-mediated mitophagy suppressed cardiomyocyte apoptosis by downregulating Bax, caspase-9, and caspase-12 and upregulating Bcl-2 and c-IAP. Using mitochondrial tracker staining and a reactive oxygen species indicator, our assays showed that Opa1-mediated mitophagy attenuated mitochondrial fission and reduced ROS production in cardiomyocytes. In addition, we found that inhibition of the MAPK/ERK pathway abolished the antioxidant action of Opa1-mediated mitophagy in these cells. Taken together, our data demonstrate that Opa1-mediated mitophagy protects cardiomyocytes against oxidative stress damage through inhibition of mitochondrial fission and activation of MAPK/ERK signaling. These findings reveal a critical role for Opa1 in the modulation of cardiomyocyte redox balance and suggest a potential target for the treatment of myocardial infarction.
Collapse
|
20
|
Anhê FF, Jensen BAH, Perazza LR, Tchernof A, Schertzer JD, Marette A. Bacterial Postbiotics as Promising Tools to Mitigate Cardiometabolic Diseases. J Lipid Atheroscler 2021; 10:123-129. [PMID: 34095007 PMCID: PMC8159759 DOI: 10.12997/jla.2021.10.2.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/12/2021] [Accepted: 01/29/2021] [Indexed: 12/25/2022] Open
Abstract
Gut microbes dictate critical features of host immunometabolism. Certain bacterial components and metabolites (termed postbiotics) mitigate cardiometabolic diseases whereas others potentiate pathological processes. In this review, we discuss key aspects related to the usefulness of bacterial-related molecules strategically positioned as promising treatment strategies for cardiometabolic diseases.
Collapse
Affiliation(s)
- Fernando F Anhê
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Benjamin A H Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lais Rossi Perazza
- Quebec Heart and Lung Research Institute, Laval University, Quebec, Canada
| | - André Tchernof
- Quebec Heart and Lung Research Institute, Laval University, Quebec, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - André Marette
- Quebec Heart and Lung Research Institute, Laval University, Quebec, Canada
| |
Collapse
|
21
|
Bai J, Liu C, Zhu P, Li Y. Novel Insights Into Molecular Mechanism of Mitochondria in Diabetic Cardiomyopathy. Front Physiol 2021; 11:609157. [PMID: 33536936 PMCID: PMC7849834 DOI: 10.3389/fphys.2020.609157] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular complication is one of the significant causes of death in diabetic mellitus (DM) in which diabetic cardiomyopathy, independent of hypertension, cardiac valvular disease, and coronary atherosclerosis, occupies an important position. Although the detailed pathogenesis of diabetic cardiomyopathy remains unclear currently, mitochondrial morphological abnormality and dysfunction were observed in diabetic cardiomyopathy animal models according to much research, suggesting that mitochondrial structural and functional impairment played an integral role in the formation of diabetic cardiomyopathy. Thus, we have summarized the effect of mitochondria on the process of diabetic cardiomyopathy, including abnormal mitochondrial morphology, mitochondrial energy metabolism disorder, enhanced mitochondrial oxidative stress, mitochondrial unbalanced calcium homeostasis, and mitochondrial autophagy. Based on the above mechanisms and the related evidence, more therapeutic strategies targeting mitochondria in diabetic cardiomyopathy have been and will be proposed to delay the progression of the disease.
Collapse
Affiliation(s)
- Jing Bai
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chuanbin Liu
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Pingjun Zhu
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yang Li
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
22
|
González-Ramos S, Fernández-García V, Recalde M, Rodríguez C, Martínez-González J, Andrés V, Martín-Sanz P, Boscá L. Deletion or Inhibition of NOD1 Favors Plaque Stability and Attenuates Atherothrombosis in Advanced Atherogenesis †. Cells 2020; 9:cells9092067. [PMID: 32927803 PMCID: PMC7564689 DOI: 10.3390/cells9092067] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Atherothrombosis, the main cause of acute coronary syndromes (ACS), is characterized by the rupture of the atherosclerotic plaque followed by the formation of thrombi. Fatal plaque rupture sites show large necrotic cores combined with high levels of inflammation and thin layers of collagen. Plaque necrosis due to the death of macrophages and smooth muscle cells (SMCs) remains critical in the process. To determine the contribution of the innate immunity receptor NOD1 to the stability of atherosclerotic plaque, Apoe-/- and Apoe-/- Nod1-/- atherosclerosis prone mice were placed on a high-fat diet for 16 weeks to assess post-mortem advanced atherosclerosis in the aortic sinus. The proliferation and apoptosis activity were analyzed, as well as the foam cell formation capacity in these lesions and in primary cultures of macrophages and vascular SMCs obtained from both groups of mice. Our results reinforce the preeminent role for NOD1 in human atherosclerosis. Advanced plaque analysis in the Apoe-/- atherosclerosis model suggests that NOD1 deficiency may decrease the risk of atherothrombosis by decreasing leukocyte infiltration and reducing macrophage apoptosis. Furthermore, Nod1-/- SMCs exhibit higher proliferation rates and decreased apoptotic activity, contributing to thicker fibrous caps with reduced content of pro-thrombotic collagen. These findings demonstrate a direct link between NOD1 and plaque vulnerability through effects on both macrophages and SMCs, suggesting promising insights for early detection of biomarkers for treating patients before ACS occurs.
Collapse
MESH Headings
- Animals
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cells, Cultured
- Gene Deletion
- Humans
- Macrophages
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle
- Nod1 Signaling Adaptor Protein/physiology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
Collapse
Affiliation(s)
- Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; (V.F.-G.); (M.R.); (P.M.-S.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (C.R.); (J.M.-G.); (V.A.)
- Correspondence: (S.G.-R.); (L.B.); Tel.: +34-(0)91-497-2747 (ext. 5345) (L.B.)
| | - Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; (V.F.-G.); (M.R.); (P.M.-S.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (C.R.); (J.M.-G.); (V.A.)
| | - Miriam Recalde
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; (V.F.-G.); (M.R.); (P.M.-S.)
| | - Cristina Rodríguez
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (C.R.); (J.M.-G.); (V.A.)
- Institut de Recerca del Hospital de la Santa Creu i Sant Pau-Programa ICCC, IIB Sant Pau, 08041 Barcelona, Spain
| | - José Martínez-González
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (C.R.); (J.M.-G.); (V.A.)
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), IIB Sant Pau, 08041 Barcelona, Spain
| | - Vicente Andrés
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (C.R.); (J.M.-G.); (V.A.)
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; (V.F.-G.); (M.R.); (P.M.-S.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; (V.F.-G.); (M.R.); (P.M.-S.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain; (C.R.); (J.M.-G.); (V.A.)
- Correspondence: (S.G.-R.); (L.B.); Tel.: +34-(0)91-497-2747 (ext. 5345) (L.B.)
| |
Collapse
|