1
|
Kupčík R, Lenčová O, Mazurová Y, Štěrba M, Vajrychová M. Methodological Aspects of μLC-MS/MS for Wide-Scale Proteomic Analysis of Anthracycline-Induced Cardiomyopathy. ACS OMEGA 2025; 10:11980-11993. [PMID: 40191338 PMCID: PMC11966270 DOI: 10.1021/acsomega.4c09377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 04/09/2025]
Abstract
The efforts to utilize microflow liquid chromatography hyphenated to tandem mass spectrometry (μLC-MS/MS) for deep-scale proteomic analysis are still growing. In this work, two-dimensional LC separation and peptide derivatization by a tandem mass tag (TMT) were used to assess the capability of μLC-MS/MS to reveal protein changes associated with the severe chronic anthracycline cardiotoxicity phenotype in comparison with nanoflow liquid chromatography (nLC-MS/MS). The analysis of the control and anthracycline-treated rabbit myocardium by μLC-MS/MS and nLC-MS/MS allowed quantification of 3956 and 4549 proteins, respectively, with 84% of these proteins shared in both data sets. Both nLC-MS/MS and μLC-MS/MS revealed marked global proteome dysregulation in severe anthracycline cardiotoxicity, with a significant change in approximately 55% of all detected proteins. The μLC-MS/MS analysis allowed less compressed and more precise determination of the TMT channel ratio and correspondingly broader fold-change protein distribution than nLC-MS/MS. The total number of significantly changed proteins was higher in nLC-MS/MS (2498 vs 2183, 1900 proteins shared), whereas the opposite was true for a number of significantly changed proteins with a fold-change cutoff ≥ 2 (535 vs 820). The profound changes concerned mainly proteins of cardiomyocyte sarcomeres, costameres, intercalated discs, mitochondria, and extracellular matrix. In addition, distinct alterations in immune and defense response were found with a remarkable involvement of type I interferon signaling that has been recently hypothesized to be essential for anthracycline cardiotoxicity pathogenesis. Hence, μLC-MS/MS was found to be a sound alternative to nLC-MS/MS that can be useful for comprehensive mapping of global myocardial proteome alterations such as those associated with severe anthracycline cardiotoxicity.
Collapse
Affiliation(s)
- Rudolf Kupčík
- Biomedical
Research Centre, University Hospital Hradec
Králové, Hradec Králové 500 05, Czech Republic
| | - Olga Lenčová
- Department
of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové 500 03, Czech Republic
| | - Yvona Mazurová
- Department
of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové 500 03, Czech Republic
| | - Martin Štěrba
- Department
of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové 500 03, Czech Republic
| | - Marie Vajrychová
- Biomedical
Research Centre, University Hospital Hradec
Králové, Hradec Králové 500 05, Czech Republic
| |
Collapse
|
2
|
Yan H, Yao W, Li Y, Li T, Song K, Yan P, Dang Y. Cardiometabolic Modulation by Semaglutide Contributes to Cardioprotection in Rats with Myocardial Infarction. Drug Des Devel Ther 2024; 18:5485-5500. [PMID: 39640291 PMCID: PMC11618856 DOI: 10.2147/dddt.s491970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Background Acute myocardial infarction (AMI) is a significant clinical challenge. Semaglutide has therapeutic potential in cardiovascular disease management, but its specific impact and mechanisms in AMI are not fully understood. Methods Twenty-four male Sprague-Dawley rats were divided into three groups: control (Control), infarction-only (MI), and semaglutide-treated (SEMA). Weight, blood glucose, and lipid profiles were analyzed. Cardiac function was evaluated via echocardiography. Histopathological assessment and immunohistochemical analysis were performed. Untargeted metabolomic analysis using LC-MS/MS was utilized. Results Semaglutide treatment was associated with a reduction in body weight, blood glucose, total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C), as well as an enhancement in the left ventricular ejection fraction (Control vs MI vs SEMA, 69.13±4.30 vs 30.16±3.17 vs 39.81±6.13, P < 0.05). It also had a lower collagen volume fraction (3.05 vs 34.05 vs 17.73, P < 0.05) and ameliorated the accumulation of glycogen in the myocardium. Metabolomic profiling revealed differentially expressed metabolites between the control/MI and MI/SEMA groups, predominantly within benzenoid, lipid, and organic acid categories. Pathway enrichment analysis highlighted amino sugar and nucleotide sugar metabolism, chlorocyclohexane and chlorobenzene degradation, and phenylalanine, tyrosine, and tryptophan biosynthesis. Random forest analysis identified key metabolites, including downregulated Docusate sodium, 1-(2-Thienyl)-1-heptanone, and Adenylyl-molybdopterin, alongside upregulated Methylenediphosphonic acid, Choline sulfate, and Lactosamine. Conclusion Semaglutide significantly ameliorated myocardial fibrosis and metabolic dysregulation in rats post-myocardial infarction. Its mechanism involves modulating glucose metabolism, lipid metabolism, and organic acid metabolism. Targeted metabolites, including Docusate sodium, 1-(2-Thienyl)-1-heptanone, Adenylyl-molybdopterin, Methylenediphosphonic acid, Choline sulfate, and Lactosamine, are implicated in the metabolic reprogramming induced by semaglutide.
Collapse
Affiliation(s)
- Haihao Yan
- Department of Internal Medicine, Graduate School of Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Wenjing Yao
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| | - Yanhong Li
- Department of Internal Medicine, Graduate School of Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
| | - Tianxing Li
- Department of Graduate School of Hebei North University, Zhangjiakou, Hebei, 075000, People’s Republic of China
| | - Kexin Song
- Department of Internal Medicine, Graduate School of Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
| | - Pan Yan
- Department of Internal Medicine, Yongnian District Traditional Chinese Medicine Hospital, Handan, Hebei, 057150, People’s Republic of China
| | - Yi Dang
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People’s Republic of China
| |
Collapse
|
3
|
Liu C, Zhang Q, Zhou H, Jin L, Liu C, Yang M, Zhao X, Ding W, Xie W, Kong H. GLP-1R activation attenuates the progression of pulmonary fibrosis via disrupting NLRP3 inflammasome/PFKFB3-driven glycolysis interaction and histone lactylation. J Transl Med 2024; 22:954. [PMID: 39434134 PMCID: PMC11492558 DOI: 10.1186/s12967-024-05753-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Pulmonary fibrosis is a serious interstitial lung disease with no viable treatment except for lung transplantation. Glucagon-like peptide-1 receptor (GLP-1R), commonly regarded as an antidiabetic target, exerts antifibrotic effects on various types of organ fibrosis. However, whether GLP-1R modulates the development and progression of pulmonary fibrosis remains unclear. In this study, we investigated the antifibrotic effect of GLP-1R using in vitro and in vivo models of pulmonary fibrosis. METHODS A silica-induced pulmonary fibrosis mouse model was established to evaluate the protective effects of activating GLP-1R with liraglutide in vivo. Primary cultured lung fibroblasts treated with TGF-β1 combined with IL-1β (TGF-β1 + IL-1β) were used to explore the specific effects of liraglutide, MCC950, and 3PO on fibroblast activation in vitro. Cell metabolism assay was performed to determine the glycolytic rate and mitochondrial respiration. RNA sequencing was utilized to analyse the underlying molecular mechanisms by which liraglutide affects fibroblast activation. ChIP‒qPCR was used to evaluate histone lactylation at the promoters of profibrotic genes in TGF-β1 + IL-1β- or exogenous lactate-stimulated lung fibroblasts. RESULTS Activating GLP-1R with liraglutide attenuated pulmonary inflammation and fibrosis in mice exposed to silica. Pharmacological inhibition of the NLRP3 inflammasome suppressed PFKFB3-driven glycolysis and vice versa, resulting in decreased lactate production in TGF-β1 + IL-1β-stimulated lung fibroblasts. Activating GLP-1R inhibited TGF-β1 + IL-1β-induced fibroblast activation by disrupting the interaction between the NLRP3 inflammasome and PFKFB3-driven glycolysis and subsequently prevented lactate-mediated histone lactylation to reduce pro-fibrotic gene expression. In addition, activating GLP-1R protected mitochondria against the TGF-β1 + IL-1β-induced increase in oxidative phosphorylation in fibroblasts. In exogenous lactate-treated lung fibroblasts, activating GLP-1R not only repressed NLRP3 inflammasome activation but also alleviated p300-mediated histone lactylation. Finally, GLP-1R activation blocked silica-treated macrophage-conditioned media-induced lung fibroblast activation. CONCLUSIONS The antifibrotic effects of GLP-1R activation on pulmonary fibrosis could be attributed to the inhibition of the interaction between NLRP3 inflammasome and PFKFB3-driven glycolysis, and histone lactylation in lung fibroblasts. Thus, GLP-1R is a specific therapeutic target for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Chenyang Liu
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China
| | - Qun Zhang
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China
| | - Hong Zhou
- Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, Jiangsu, 214023, P. R. China
| | - Linling Jin
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China
| | - Chang Liu
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China
| | - Mingxia Yang
- Department of Pulmonary & Critical Care Medicine, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213003, P. R. China
| | - Xinyun Zhao
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China
| | - Wenqiu Ding
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China
| | - Weiping Xie
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China.
| | - Hui Kong
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China.
| |
Collapse
|
4
|
Karpov OA, Stotland A, Raedschelders K, Chazarin B, Ai L, Murray CI, Van Eyk JE. Proteomics of the heart. Physiol Rev 2024; 104:931-982. [PMID: 38300522 PMCID: PMC11381016 DOI: 10.1152/physrev.00026.2023] [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: 07/03/2023] [Revised: 12/25/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
Mass spectrometry-based proteomics is a sophisticated identification tool specializing in portraying protein dynamics at a molecular level. Proteomics provides biologists with a snapshot of context-dependent protein and proteoform expression, structural conformations, dynamic turnover, and protein-protein interactions. Cardiac proteomics can offer a broader and deeper understanding of the molecular mechanisms that underscore cardiovascular disease, and it is foundational to the development of future therapeutic interventions. This review encapsulates the evolution, current technologies, and future perspectives of proteomic-based mass spectrometry as it applies to the study of the heart. Key technological advancements have allowed researchers to study proteomes at a single-cell level and employ robot-assisted automation systems for enhanced sample preparation techniques, and the increase in fidelity of the mass spectrometers has allowed for the unambiguous identification of numerous dynamic posttranslational modifications. Animal models of cardiovascular disease, ranging from early animal experiments to current sophisticated models of heart failure with preserved ejection fraction, have provided the tools to study a challenging organ in the laboratory. Further technological development will pave the way for the implementation of proteomics even closer within the clinical setting, allowing not only scientists but also patients to benefit from an understanding of protein interplay as it relates to cardiac disease physiology.
Collapse
Affiliation(s)
- Oleg A Karpov
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Aleksandr Stotland
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Koen Raedschelders
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Blandine Chazarin
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Lizhuo Ai
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Christopher I Murray
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| |
Collapse
|
5
|
Ma YL, Kong CY, Guo Z, Wang MY, Wang P, Liu FY, Yang D, Yang Z, Tang QZ. Semaglutide ameliorates cardiac remodeling in male mice by optimizing energy substrate utilization through the Creb5/NR4a1 axis. Nat Commun 2024; 15:4757. [PMID: 38834564 PMCID: PMC11150406 DOI: 10.1038/s41467-024-48970-2] [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: 01/10/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024] Open
Abstract
Semaglutide, a glucagon-like peptide-1 receptor agonist, is clinically used as a glucose-lowering and weight loss medication due to its effects on energy metabolism. In heart failure, energy production is impaired due to altered mitochondrial function and increased glycolysis. However, the impact of semaglutide on cardiomyocyte metabolism under pressure overload remains unclear. Here we demonstrate that semaglutide improves cardiac function and reduces hypertrophy and fibrosis in a mouse model of pressure overload-induced heart failure. Semaglutide preserves mitochondrial structure and function under chronic stress. Metabolomics reveals that semaglutide reduces mitochondrial damage, lipid accumulation, and ATP deficiency by promoting pyruvate entry into the tricarboxylic acid cycle and increasing fatty acid oxidation. Transcriptional analysis shows that semaglutide regulates myocardial energy metabolism through the Creb5/NR4a1 axis in the PI3K/AKT pathway, reducing NR4a1 expression and its translocation to mitochondria. NR4a1 knockdown ameliorates mitochondrial dysfunction and abnormal glucose and lipid metabolism in the heart. These findings suggest that semaglutide may be a therapeutic agent for improving cardiac remodeling by modulating energy metabolism.
Collapse
Affiliation(s)
- Yu-Lan Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Chun-Yan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Pan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China.
| |
Collapse
|
6
|
Zhang JW, Huang LY, Li YN, Tian Y, Yu J, Wang XF. Mitochondrial carrier homolog 2 increases malignant phenotype of human gastric epithelial cells and promotes proliferation, invasion, and migration of gastric cancer cells. World J Gastrointest Oncol 2024; 16:991-1005. [PMID: 38577443 PMCID: PMC10989370 DOI: 10.4251/wjgo.v16.i3.991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/21/2023] [Accepted: 01/19/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND The precise role of mitochondrial carrier homolog 2 (MTCH2) in promoting malignancy in gastric mucosal cells and its involvement in gastric cancer cell metastasis have not been fully elucidated. AIM To determine the role of MTCH2 in gastric cancer. METHODS We collected 65 samples of poorly differentiated gastric cancer tissue and adjacent tissues, constructed MTCH2-overexpressing and MTCH2-knockdown cell models, and evaluated the proliferation, migration, and invasion of human gastric epithelial cells (GES-1) and human gastric cancer cells (AGS) cells. The mitochondrial membrane potential (MMP), mitochondrial permeability transformation pore (mPTP) and ATP fluorescence probe were used to detect mitochondrial function. Mitochondrial function and ATP synthase protein levels were detected via Western blotting. RESULTS The expression of MTCH2 and ATP2A2 in gastric cancer tissues was significantly greater than that in adjacent tissues. Overexpression of MTCH2 promoted colony formation, invasion, migration, MMP expression and ATP production in GES-1 and AGS cells while upregulating ATP2A2 expression and inhibiting cell apoptosis; knockdown of MTCH2 had the opposite effect, promoting overactivation of the mPTP and promoting apoptosis. CONCLUSION MTCH2 can increase the malignant phenotype of GES-1 cells and promote the proliferation, invasion, and migration of gastric cancer cells by regulating mitochondrial function, providing a basis for targeted therapy for gastric cancer cells.
Collapse
Affiliation(s)
- Jing-Wen Zhang
- School of Basic Medical Science, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling-Yan Huang
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ya-Ning Li
- School of Basic Medical Science, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ying Tian
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jia Yu
- School of Basic Medical Science, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Xiao-Fei Wang
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei Province, China
| |
Collapse
|
7
|
A Molecular (Not Very Becoming) Picture of Stressed Arteries and Heart, with Some Therapeutic Hope. Int J Mol Sci 2023; 24:ijms24043870. [PMID: 36835280 PMCID: PMC9968135 DOI: 10.3390/ijms24043870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
This Special Issue has focused on molecular mechanisms (vascular calcification, endothelial dysfunction, cardiac remodelling, inflammation, oxidative stress, etc [...].
Collapse
|
8
|
Pietras Ł, Stefanik E, Rakus D, Gizak A. FBP2–A New Player in Regulation of Motility of Mitochondria and Stability of Microtubules in Cardiomyocytes. Cells 2022; 11:cells11101710. [PMID: 35626746 PMCID: PMC9139521 DOI: 10.3390/cells11101710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
Recently, we have shown that the physiological roles of a multifunctional protein fructose 1,6-bisphosphatase 2 (FBP2, also called muscle FBP) depend on the oligomeric state of the protein. Here, we present several lines of evidence that in HL-1 cardiomyocytes, a forced, chemically induced reduction in the FBP2 dimer-tetramer ratio that imitates AMP and NAD+ action and restricts FBP2-mitochondria interaction, results in an increase in Tau phosphorylation, augmentation of FBP2-Tau and FBP2-MAP1B interactions, disturbance of tubulin network, marked reduction in the speed of mitochondrial trafficking and increase in mitophagy. These results not only highlight the significance of oligomerization for the regulation of FBP2 physiological role in the cell, but they also demonstrate a novel, important cellular function of this multitasking protein—a function that might be crucial for processes that take place during physiological and pathological cardiac remodeling, and during the onset of diseases which are rooted in the destabilization of MT and/or mitochondrial network dynamics.
Collapse
|
9
|
Cao Y, Ma W, Liu Z, Pei Y, Zhu Y, Chen F, Zou L, Jiang Y, Liu X, Huang J, Xiao W, Han X. Early predictive value of platelet function for clinical outcome in sepsis. J Infect 2022; 84:628-636. [PMID: 35150766 DOI: 10.1016/j.jinf.2022.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/31/2022] [Accepted: 02/07/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Sepsis is the leading course of morbidity and mortality in critically ill patients. This study aimed to evaluate the predictive value of the platelet aggregation for mortality in patients with sepsis. In addition, the relationship between impaired mitochondria and the platelet aggregation was explored. METHOD This was a prospective, observational cohort study. The platelet aggregation rate in response to adenosine diphosphate (ADP) was assessed. The primary outcome was 28-day mortality. Platelet mitochondrial parameters, including adenosine triphosphate(ATP), mitochondrial membrane potential (MMP) and mitochondrial permeability transition pore (mPTP) opening, were measured. Platelet mitochondrial ultrastructure was observed using transmission electron microscopy. RESULTS 86 patients with 65 survivors and 21 non-survivors were enrolled. Platelets of non-survivors with sepsis were hyporesponsive to ADP, in terms of maximal aggregation rate (P < 0.001). Receiver operating characteristic curves analysis demonstrated that the AUC estimated 28-day mortality for platelet aggregation rate was 0.814. At the optimal cut-off value of 35.8% for platelet aggregation rate, the sensitivity was 86.2% and the specificity was 66.7%. Kaplan-Meier analysis showed that a platelet aggregation rate of less than 35.8% was associated closely with poor survival. After adjusting for lactate by Cox regression analysis, platelet aggregation rate was identified as an independent predictor for the probability of 28-day mortality. Compared with survivors, non-survivors showed a significant reduction in platelet ATP and MMP-index (both P < 0.001), and a remarkable increase in mPTP opening (P < 0.001). ATP and MMP-index were positively correlated with platelet aggregation rate (R square=0.75, R square=0.44, respectively). CONCLUSION Platelet aggregation rate could be an early predictive biomarker for mortality in sepsis. Impaired platelet mitochondrial activity affects platelet aggregation and correlates with the severity of sepsis.
Collapse
Affiliation(s)
- Yan Cao
- Department of Emergency, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61 Western Jiefang Road, Changsha, Hunan 410005, China
| | - Wenfeng Ma
- Department of Emergency, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Changsha, China
| | - Zhengyu Liu
- Department of Cardiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China; Clinical Research Center for Heart Failure of Hunan Province, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yanfang Pei
- Department of Emergency, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61 Western Jiefang Road, Changsha, Hunan 410005, China
| | - Yimin Zhu
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Fang Chen
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Lianhong Zou
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yu Jiang
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Xiehong Liu
- Institute of Emergency Medicine, Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Jie Huang
- Department of Emergency, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61 Western Jiefang Road, Changsha, Hunan 410005, China
| | - Weiwei Xiao
- Department of Emergency, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61 Western Jiefang Road, Changsha, Hunan 410005, China.
| | - Xiaotong Han
- Department of Emergency, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61 Western Jiefang Road, Changsha, Hunan 410005, China.
| |
Collapse
|