1
|
Li Z, Xu P, Deng Y, Duan R, Peng Q, Wang S, Xu Z, Hong Y, Zhang Y. M1 Microglia-Derived Exosomes Promote A1 Astrocyte Activation and Aggravate Ischemic Injury via circSTRN3/miR-331-5p/MAVS/NF-κB Pathway. J Inflamm Res 2024; 17:9285-9305. [PMID: 39588134 PMCID: PMC11587797 DOI: 10.2147/jir.s485252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024] Open
Abstract
Background After ischemic stroke (IS), microglia and astrocytes undergo polarization, transforming into a pro-inflammatory phenotype (M1 or A1). According to previous studies, exosomes might play an important role in the interplay between M1 microglia and A1 astrocytes after IS. Methods We used the microglial oxygen-glucose deprivation/reperfusion (OGD/R) model and ultracentrifugation to extract M1 microglial exosomes (M1-exos). Subsequently, we identified circSTRN3 enriched in exosomes through RNA sequencing and detected the role of circSTRN3 in astrocyte activation based on bioinformatics analysis, immunofluorescence, Western blotting, and polymerase chain reaction analysis. We validated these findings in the middle cerebral artery occlusion/reperfusion (MCAO/R) model of adult male C57BL/6J mice. Finally, we confirmed the correlation among circSTRN3, miR-331-5p, and stroke severity score in exosomes isolated from peripheral blood of IS patients. Results Our findings revealed that M1-exos promoted A1 astrocyte activation. CircSTRN3 was abundant in M1-exos, which could sponge miR-331-5p to affect mitochondrial antiviral signaling protein (MAVS), activate NF-κB pathway, and participate in A1 astrocyte activation. In addition, overexpressed circSTRN3 augmented the infarct size and neurological dysfunction in MCAO/R models, while miR-331-5p mimics reversed the effect. Furthermore, circSTRN3 in IS patients was positively correlated with stroke severity score (R 2 = 0.83, P < 0.001), while miR-331-5p demonstrated a negative correlation with the same score (R 2 = 0.81, P < 0.001). Conclusion Taken together, our research indicated that circSTRN3 from M1-exos could promote A1 astrocyte activation and exacerbate ischemic brain injury via miR331-5p/MAVS/NF-κB axis.
Collapse
Affiliation(s)
- Zhongyuan Li
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210000, People’s Republic of China
| | - Pengfei Xu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, People’s Republic of China
| | - Yang Deng
- Department of Neurology, Nanjing First Hospital, China Pharmaceutical University, Nanjing, 210006, People’s Republic of China
| | - Rui Duan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210000, People’s Republic of China
| | - Qiang Peng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210000, People’s Republic of China
| | - Shiyao Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210000, People’s Republic of China
| | - Zhaohan Xu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210000, People’s Republic of China
| | - Ye Hong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210000, People’s Republic of China
| | - Yingdong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210000, People’s Republic of China
| |
Collapse
|
2
|
Wang Z, Wojciechowicz M, Rosen J, Elmas A, Song WM, Liu Y, Huang KL. Master regulators governing protein abundance across ten human cancer types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.619147. [PMID: 39605415 PMCID: PMC11601414 DOI: 10.1101/2024.11.11.619147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Protein abundance correlates only moderately with mRNA levels, and are modulated post-transcriptionally by a network of regulators including ribosomes, RNA-binding proteins (RBPs), and the proteasome. Here, we identified Master Protein abundance Regulators (MaPRs) across ten cancer types by devising a new computational pipeline that jointly analyzed transcriptomes and proteomes from 1,305 tumor samples. We identified 232 to 1,394 MaPRs per cancer type, mediating up to 79% of post-transcriptional regulatory networks. MaPRs exhibit high network connectivity, strong genetic dependency in cancer cells, and significant enrichment for RBPs. Combining tumor up-regulation, druggability, and target network analyses identified cancer-specific vulnerabilities. MaPRs predict tumor proteomic subtypes more accurately than other proteins. Finally, significant portions of RBP MaPR-target relationships were validated by experimental evidence from eCLIP binding and knockdown assays. Our findings uncover central MaPRs that govern post-transcriptional networks, highlighting diverse processes underlying human proteome regulation and identifying key regulators in cancer biology.
Collapse
Affiliation(s)
- Zishan Wang
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Megan Wojciechowicz
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Jordan Rosen
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Abdulkadir Elmas
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Won-Min Song
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Yansheng Liu
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Kuan-lin Huang
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| |
Collapse
|
3
|
Pei H, Qu J, Chen J, Zhao G, Lu Z. S100A9 as a Key Myocardial Injury Factor Interacting with ATP5 Exacerbates Mitochondrial Dysfunction and Oxidative Stress in Sepsis-Induced Cardiomyopathy. J Inflamm Res 2024; 17:4483-4503. [PMID: 39006491 PMCID: PMC11246037 DOI: 10.2147/jir.s457340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Purpose Sepsis-induced cardiomyopathy (SICM) is a prevalent cardiac dysfunction caused by sepsis. Mitochondrial dysfunction is a crucial pathogenic factor associated with adverse cardiovascular adverse events; however, research on SICM remains insufficient. Methods To investigate the factors contributing to the pathological progression of SICM, we performed a comprehensive analysis of transcriptomic data from the GEO database using bioinformatics and machine learning techniques. CRISPR-Cas9 S100A9 knockout mice and primary cardiomyocytes were exposed to lipopolysaccharide to simulate SICM. Transcriptome analysis and mass spectrometry of primary cardiomyocytes were used to determine the potential pathogenic mechanisms of S100A9. The mitochondrial ultrastructure and mitochondrial membrane potential (MMP) were detected using transmission electron microscopy and flow cytometry, respectively. Pink1/Parkin and Drp1 proteins were detected using Western blotting to evaluate mitochondrial autophagy and division. The mtDNA and mRNA levels of mitochondrial transcription factors and synthases were evaluated using real-time polymerase chain reaction. Results Bioinformatics analysis identified 12 common differentially expressed genes, including SERPINA3N, LCN2, MS4A6D, LRG1, OSMR, SOCS3, FCGR2b, S100A9, S100A8, CASP4, ABCA8A, and NFKBIZ. Significant S100A9 upregulation was closely associated with myocardial injury exacerbation and cardiac function deterioration. GSEA revealed that myocardial contractile function, oxidative stress, and mitochondrial function were significantly affected by S100A9. Knocking out S100A9 alleviates the inflammatory response and mitochondrial dysfunction. The interaction of S100A9 with ATP5 enhanced mitochondrial division and autophagy, inhibited MMP and ATP synthesis, and induced oxidative stress, which are related to the Nlrp3-Nfkb-Caspase1 and Drp1-Pink1-Parkin signaling pathways. The expression of mitochondrial transcription factors (TFAM and TFBM) and ATP synthetases (ATP6 and ATP8, as well as COX1, COX2, and COX3) was further suppressed by S100A9 in SICM. Targeted S100A9 inhibition by paquinimod partially reversed myocardial mitochondrial dysfunction and oxidative stress. Conclusion The interaction of S100A9 with ATP5 exacerbates myocardial damage in sepsis by inducing mitochondrial dysfunction and oxidative stress.
Collapse
Affiliation(s)
- Hui Pei
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Jie Qu
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Jianming Chen
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Guangju Zhao
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - ZhongQiu Lu
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, People’s Republic of China
| |
Collapse
|
4
|
Hinton AO, N'jai AU, Vue Z, Wanjalla C. Connection Between HIV and Mitochondria in Cardiovascular Disease and Implications for Treatments. Circ Res 2024; 134:1581-1606. [PMID: 38781302 PMCID: PMC11122810 DOI: 10.1161/circresaha.124.324296] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
HIV infection and antiretroviral therapy alter mitochondrial function, which can progressively lead to mitochondrial damage and accelerated aging. The interaction between persistent HIV reservoirs and mitochondria may provide insight into the relatively high rates of cardiovascular disease and mortality in persons living with HIV. In this review, we explore the intricate relationship between HIV and mitochondrial function, highlighting the potential for novel therapeutic strategies in the context of cardiovascular diseases. We reflect on mitochondrial dynamics, mitochondrial DNA, and mitochondrial antiviral signaling protein in the context of HIV. Furthermore, we summarize how toxicities related to early antiretroviral therapy and current highly active antiretroviral therapy can contribute to mitochondrial dysregulation, chronic inflammation, and poor clinical outcomes. There is a need to understand the mechanisms and develop new targeted therapies. We further consider current and potential future therapies for HIV and their interplay with mitochondria. We reflect on the next-generation antiretroviral therapies and HIV cure due to the direct and indirect effects of HIV persistence, associated comorbidities, coinfections, and the advancement of interdisciplinary research fields. This includes exploring novel and creative approaches to target mitochondria for therapeutic intervention.
Collapse
Affiliation(s)
- Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (A.O.H., Z.V.)
| | - Alhaji U N'jai
- Biological Sciences, Fourah Bay College and College of Medicine and Allied Health Sciences (COMAHS), University of Sierra Leone, Freetown, Sierra Leone and Koinadugu College, Kabala (A.U.N.)
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (A.O.H., Z.V.)
| | - Celestine Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.W.)
| |
Collapse
|
5
|
Tran TAT, Iwata Y, Hoang LT, Kitajima S, Yoneda-Nakagawa S, Oshima M, Sakai N, Toyama T, Yamamura Y, Yamazaki H, Hara A, Shimizu M, Sako K, Minami T, Yuasa T, Horikoshi K, Hayashi D, Kajikawa S, Wada T. Protective Role of MAVS Signaling for Murine Lipopolysaccharide-Induced Acute Kidney Injury. Immunohorizons 2024; 8:1-18. [PMID: 38169549 PMCID: PMC10835654 DOI: 10.4049/immunohorizons.2300069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 01/05/2024] Open
Abstract
Despite treatment advances, acute kidney injury (AKI)-related mortality rates are still high in hospitalized adults, often due to sepsis. Sepsis and AKI could synergistically worsen the outcomes of critically ill patients. TLR4 signaling and mitochondrial antiviral signaling protein (MAVS) signaling are innate immune responses essential in kidney diseases, but their involvement in sepsis-associated AKI (SA-AKI) remains unclear. We studied the role of MAVS in kidney injury related to the TLR4 signaling pathway using a murine LPS-induced AKI model in wild-type and MAVS-knockout mice. We confirmed the importance of M1 macrophage in SA-AKI through in vivo assessment of inflammatory responses. The TLR4 signaling pathway was upregulated in activated bone marrow-derived macrophages, in which MAVS helped maintain the LPS-suppressed TLR4 mRNA level. MAVS regulated redox homeostasis via NADPH oxidase Nox2 and mitochondrial reverse electron transport in macrophages to alleviate the TLR4 signaling response to LPS. Hypoxia-inducible factor 1α (HIF-1α) and AP-1 were key regulators of TLR4 transcription and connected MAVS-dependent reactive oxygen species signaling with the TLR4 pathway. Inhibition of succinate dehydrogenase could partly reduce inflammation in LPS-treated bone marrow-derived macrophages without MAVS. These findings highlight the renoprotective role of MAVS in LPS-induced AKI by regulating reactive oxygen species generation-related genes and maintaining redox balance. Controlling redox homeostasis through MAVS signaling may be a promising therapy for SA-AKI.
Collapse
Affiliation(s)
- Trang Anh Thi Tran
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Yasunori Iwata
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
- Division of Infection Control, Kanazawa University Hospital, Kanazawa, Japan
| | - Linh Thuy Hoang
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Shinji Kitajima
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
- Division of Blood Purification, Kanazawa University Hospital, Kanazawa, Japan
| | | | - Megumi Oshima
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Norihiko Sakai
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
- Division of Blood Purification, Kanazawa University Hospital, Kanazawa, Japan
| | - Tadashi Toyama
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Yuta Yamamura
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Hiroka Yamazaki
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Akinori Hara
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Miho Shimizu
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Keisuke Sako
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Taichiro Minami
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Takahiro Yuasa
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Keisuke Horikoshi
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Daiki Hayashi
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Sho Kajikawa
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Takashi Wada
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
6
|
Liu Y, Wang D, Jin Y, Sun G, Lou Q, Wang H, Li W. Costunolide ameliorates angiotensin II-induced atrial inflammation and fibrosis by regulating mitochondrial function and oxidative stress in mice: A possible therapeutic approach for atrial fibrillation. Microvasc Res 2024; 151:104600. [PMID: 37666318 DOI: 10.1016/j.mvr.2023.104600] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
Atrial fibrillation (AF) is a cardiac disease characterized by disordered atrial electrical activity. Atrial inflammation and fibrosis are involved in AF progression. Costunolide (COS) is a sesquiterpene lactone containing anti-inflammatory and anti-fibrotic activities. This study aims to explore the underlying mechanisms by which COS protects against AF. Male C57BL/6 mice (8- to 10-week-old) were infused with angiotensin (Ang) II for 3 weeks. Meanwhile, different doses of COS (COS-L: 10 mg/kg, COS-H: 20 mg/kg) were administered to mice by intragastric treatment. The results showed irregular and rapid heart rates in Ang II-treated mice. Moreover, the levels of inflammatory cytokines and fibrotic factors were elevated in mice. COS triggered a reduction of Ang II-induced inflammation and fibrosis, which conferred a protective effect. Mechanistically, mitochondrial dysfunction with mitochondrial respiration inhibition and aberrant ATP levels were observed after Ang II treatment. Moreover, Ang-II-induced excessive reactive oxygen species caused oxidative stress, which was further aggravated by inhibiting Nrf2 nuclear translocation. Importantly, COS diminished these Ang-II-mediated effects in mice. In conclusion, COS attenuated inflammation and fibrosis in Ang-II-treated mice by alleviating mitochondrial dysfunction and oxidative stress. Our findings represent a potential therapeutic option for AF treatment.
Collapse
Affiliation(s)
- Yushu Liu
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Dong Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, PR China
| | - Yimin Jin
- Department of General Practice, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Guifang Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Qi Lou
- Graduate Student, Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Hong Wang
- Graduate Student, Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Weimin Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China.
| |
Collapse
|
7
|
Menezes Júnior ADS, de França-e-Silva ALG, de Oliveira JM, da Silva DM. Developing Pharmacological Therapies for Atrial Fibrillation Targeting Mitochondrial Dysfunction and Oxidative Stress: A Scoping Review. Int J Mol Sci 2023; 25:535. [PMID: 38203704 PMCID: PMC10779389 DOI: 10.3390/ijms25010535] [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: 11/18/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Atrial fibrillation (AF) is a cardiac arrhythmia caused by electrophysiological anomalies in the atrial tissue, tissue degradation, structural abnormalities, and comorbidities. A direct relationship exists between AF and altered mitochondrial activity resulting from membrane potential loss, contractile dysfunction, or decreased ATP levels. This review aimed to elucidate the role of mitochondrial oxidative mechanisms in AF pathophysiology, the impact of mitochondrial oxidative stress on AF initiation and perpetuation, and current therapies. This review followed the Preferred Reporting Items for Systematic Reviews and the Meta-Analysis Extension for Scoping Reviews. PubMed, Excerpta Medica Database, and Scopus were explored until June 2023 using "MESH terms". Bibliographic references to relevant papers were also included. Oxidative stress is an imbalance that causes cellular damage from excessive oxidation, resulting in conditions such as AF. An imbalance in reactive oxygen species production and elimination can cause mitochondrial damage, cellular apoptosis, and cardiovascular diseases. Oxidative stress and inflammation are intrinsically linked, and inflammatory pathways are highly correlated with the occurrence of AF. AF is an intricate cardiac condition that requires innovative therapeutic approaches. The involvement of mitochondrial oxidative stress in the pathophysiology of AF introduces novel strategies for clinical treatment.
Collapse
Affiliation(s)
- Antônio da Silva Menezes Júnior
- Internal Medicine Department, Medicine School, Federal University of Goiás, Goiânia 74605-020, GO, Brazil; (A.L.G.d.F.-e.-S.); (D.M.d.S.)
- Medicine Department, Medical and Life School, Pontifical Catholic University of Goiás, Avenida Universitária, 1440, Sector Universitario, Goiânia 74605-010, GO, Brazil;
| | - Ana Luísa Guedes de França-e-Silva
- Internal Medicine Department, Medicine School, Federal University of Goiás, Goiânia 74605-020, GO, Brazil; (A.L.G.d.F.-e.-S.); (D.M.d.S.)
| | - Joyce Monteiro de Oliveira
- Medicine Department, Medical and Life School, Pontifical Catholic University of Goiás, Avenida Universitária, 1440, Sector Universitario, Goiânia 74605-010, GO, Brazil;
| | - Daniela Melo da Silva
- Internal Medicine Department, Medicine School, Federal University of Goiás, Goiânia 74605-020, GO, Brazil; (A.L.G.d.F.-e.-S.); (D.M.d.S.)
| |
Collapse
|
8
|
Yan S, Huang Z, Chen X, Chen H, Yang X, Gao M, Zhang X. Metabolic profiling of urinary exosomes for systemic lupus erythematosus discrimination based on HPL-SEC/MALDI-TOF MS. Anal Bioanal Chem 2023; 415:6411-6420. [PMID: 37644324 DOI: 10.1007/s00216-023-04916-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/01/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease which leads to the formation of immune complex deposits in multiple organs and has heterogeneous clinical manifestations. Currently, exosomes for liquid biopsy have been applied in diagnosis and monitoring of diseases, whereas SLE discrimination based on exosomes at the metabolic level is rarely reported. Herein, we constructed a protocol for metabolomic study of urinary exosomes from SLE patients and healthy controls (HCs) with high efficiency and throughput. Exosomes were first obtained by high-performance liquid size-exclusion chromatography (HPL-SEC), and then metabolic fingerprints of urinary exosomes were extracted by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) with high throughput and high efficency. With the statistical analysis by orthogonal partial least-squares discriminant analysis (OPLS-DA) model, SLE patients were efficiently distinguished from HCs, the area under the curve (AUC) of the receiver characteristic curve (ROC) was 1.00, and the accuracy of the unsupervised clustering heatmap was 90.32%. In addition, potential biomarkers and related metabolic pathways were analyzed. This method, with the characteristics of high throughput, high efficiency, and high accuracy, will provide the broad prospect of exosome-driven precision medicine and large-scale screening in clinical applications.
Collapse
Affiliation(s)
- Shaohan Yan
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200438, China
| | - Zhongzhou Huang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaofei Chen
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200438, China
| | - Haolin Chen
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200438, China
| | - Xue Yang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Mingxia Gao
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200438, China.
| | - Xiangmin Zhang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200438, China
| |
Collapse
|
9
|
Wu M, Pei Z, Long G, Chen H, Jia Z, Xia W. Mitochondrial antiviral signaling protein: a potential therapeutic target in renal disease. Front Immunol 2023; 14:1266461. [PMID: 37901251 PMCID: PMC10602740 DOI: 10.3389/fimmu.2023.1266461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023] Open
Abstract
Mitochondrial antiviral signaling protein (MAVS) is a key innate immune adaptor on the outer mitochondrial membrane that acts as a switch in the immune signal transduction response to viral infections. Some studies have reported that MAVS mediates NF-κB and type I interferon signaling during viral infection and is also required for optimal NLRP3 inflammasome activity. Recent studies have reported that MAVS is involved in various cancers, systemic lupus erythematosus, kidney diseases, and cardiovascular diseases. Herein, we summarize the structure, activation, pathophysiological roles, and MAVS-based therapies for renal diseases. This review provides novel insights into MAVS's role and therapeutic potential in the pathogenesis of renal diseases.
Collapse
Affiliation(s)
- Meng Wu
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiyin Pei
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Guangfeng Long
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Hongbing Chen
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Weiwei Xia
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|