1
|
Jin L, Han Z, Mao X, Lu J, Yan B, Lu Y, Liang L, Wang L, Yu Y, Sun K. Genome-wide profiling of angiogenic cis-regulatory elements unravels cis-regulatory SNPs for vascular abnormality. Sci Data 2024; 11:467. [PMID: 38719891 PMCID: PMC11078952 DOI: 10.1038/s41597-024-03272-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
Angiogenesis is extensively involved in embryonic development and requires complex regulation networks, whose defects can cause a variety of vascular abnormalities. Cis-regulatory elements control gene expression at all developmental stages, but they have not been studied or profiled in angiogenesis yet. In this study, we exploited public DNase-seq and RNA-seq datasets from a VEGFA-stimulated in vitro angiogenic model, and carried out an integrated analysis of the transcriptome and chromatin accessibility across the entire process. Totally, we generated a bank of 47,125 angiogenic cis-regulatory elements with promoter (marker by H3K4me3) and/or enhancer (marker by H3K27ac) activities. Motif enrichment analysis revealed that these angiogenic cis-regulatory elements interacted preferentially with ETS family TFs. With this tool, we performed an association study using our WES data of TAPVC and identified rs199530718 as a cis-regulatory SNP associated with disease risk. Altogether, this study generated a genome-wide bank of angiogenic cis-regulatory elements and illustrated its utility in identifying novel cis-regulatory SNPs for TAPVC, expanding new horizons of angiogenesis as well as vascular abnormality genetics.
Collapse
Affiliation(s)
- Lihui Jin
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China.
| | - Zhenyuan Han
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Xiaotong Mao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jieru Lu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
- Department of Pediatrics, Lishui People's Hospital, Lishui, 323050, China
| | - Bingqian Yan
- Department of NICU, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Yiwen Lu
- Department of Laboratory Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Lili Liang
- Department of Pediatric Endocrinology/Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Lin Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Yu Yu
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China.
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
2
|
Martínez González Á, Llópiz Castedo J, Rodeiro Escobar P, González Nunes M, Fernández López B, García Cardoner MDLÁ, Fraile Amador FJ, Rodriguez Zorrilla S, Martínez González MI, Rodeiro Marta SE. [Effectiveness of immunonutrition in the perioperative nutritional management of gastric cancer]. NUTR HOSP 2024; 41:330-337. [PMID: 38328956 DOI: 10.20960/nh.04934] [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] [Indexed: 02/09/2024] Open
Abstract
Introduction Objective: to assess the effectiveness of immunonutrition (IN) compared to standard nutritional formulas in patients undergoing gastric cancer surgery. Material and methods: this is a real-life, observational retrospective cohort study. It included 134 patients, all of whom underwent gastrectomy at Montecelo Hospital between December 2019 and December 2022. Group A (N = 79 patients) received standard nutrition, and Group B (N = 55 patients) received formulas containing arginine, nucleotides, omega-3 fatty acids, and extra virgin olive oil. This protocol was carried out both pre and postoperatively for an average period of 10 days. The study evaluated hospital stay, the need for parenteral nutrition (PN), postoperative complications, as well as anthropometric and laboratory variables. Statistical analyses were performed using Stata 16.1.® Results: in the IN group compared to the standard nutrition group, the hospital stay was reduced by 34 % (p < 0.001). The number of patients requiring PN decreased by 21.1 % (p = 0.022), and its duration also decreased by 33.2 % (p < 0.001). The risk of infectious complications was lower with IN, specifically 70.1 % less (p < 0.001). As for other postoperative complications, IN reduced the risk of intestinal obstruction by 84 % (p < 0.002), suture dehiscence by 90.9 % (p < 0.001), blood transfusion by 99.8 % (p < 0.001), pleural effusion by 90.9 % (p = 0.021), acute renal failure by 84.02 % (p = 0.047), and surgical re-intervention by 69.93 % (p < 0.011). In the IN group, there was less weight loss (p = 0.048) and a smaller decrease in postoperative albumin (p = 0.005) and cholesterol (p < 0.001). Conclusion: immunonutrition reduces postoperative complications, decreases hospital stay, and optimizes nutritional outcomes.
Collapse
|
3
|
Pant T, Uche N, Juric M, Zielonka J, Bai X. Regulation of immunomodulatory networks by Nrf2-activation in immune cells: Redox control and therapeutic potential in inflammatory diseases. Redox Biol 2024; 70:103077. [PMID: 38359749 PMCID: PMC10877431 DOI: 10.1016/j.redox.2024.103077] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024] Open
Abstract
Inflammatory diseases present a serious health challenge due to their widespread prevalence and the severe impact on patients' lives. In the quest to alleviate the burden of these diseases, nuclear factor erythroid 2-related factor 2 (Nrf2) has emerged as a pivotal player. As a transcription factor intimately involved in cellular defense against metabolic and oxidative stress, Nrf2's role in modulating the inflammatory responses of immune cells has garnered significant attention. Recent findings suggest that Nrf2's ability to alter the redox status of cells underlies its regulatory effects on immune responses. Our review delves into preclinical and clinical evidence that underscores the complex influence of Nrf2 activators on immune cell phenotypes, particularly in the inflammatory milieu. By offering a detailed analysis of Nrf2's role in different immune cell populations, we cast light on the potential of Nrf2 activators in shaping the immune response towards a more regulated state, mitigating the adverse effects of inflammation through modeling redox status of immune cells. Furthermore, we explore the innovative use of nanoencapsulation techniques that enhance the delivery and efficacy of Nrf2 activators, potentially advancing the treatment strategies for inflammatory ailments. We hope this review will stimulate the development and expansion of Nrf2-targeted treatments that could substantially improve outcomes for patients suffering from a broad range of inflammatory diseases.
Collapse
Affiliation(s)
- Tarun Pant
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA; Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Nnamdi Uche
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matea Juric
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
4
|
Sun Q, Gao J, An R, Wang M, Wang Y. Probing molecular pathways: Illuminating the connection between COVID-19 and Alzheimer's disease through the endocannabinoid system dynamics. J Med Virol 2024; 96:e29590. [PMID: 38619024 DOI: 10.1002/jmv.29590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/29/2024] [Accepted: 03/26/2024] [Indexed: 04/16/2024]
Abstract
Our study investigates the molecular link between COVID-19 and Alzheimer's disease (AD). We aim to elucidate the mechanisms by which COVID-19 may influence the onset or progression of AD. Using bioinformatic tools, we analyzed gene expression datasets from the Gene Expression Omnibus (GEO) database, including GSE147507, GSE12685, and GSE26927. Intersection analysis was utilized to identify common differentially expressed genes (CDEGs) and their shared biological pathways. Consensus clustering was conducted to group AD patients based on gene expression, followed by an analysis of the immune microenvironment and variations in shared pathway activities between clusters. Additionally, we identified transcription factor-binding sites shared by CDEGs and genes in the common pathway. The activity of the pathway and the expression levels of the CDEGs were validated using GSE164805 and GSE48350 datasets. Six CDEGs (MAL2, NECAB1, SH3GL2, EPB41L3, MEF2C, and NRGN) were identified, along with a downregulated pathway, the endocannabinoid (ECS) signaling pathway, common to both AD and COVID-19. These CDEGs showed a significant correlation with ECS activity (p < 0.05) and immune functions. The ECS pathway was enriched in healthy individuals' brains and downregulated in AD patients. Validation using GSE164805 and GSE48350 datasets confirmed the differential expression of these genes in COVID-19 and AD tissues. Our findings reveal a potential pathogenetic link between COVID-19 and AD, mediated by CDEGs and the ECS pathway. However, further research and multicenter evidence are needed to translate these findings into clinical applications.
Collapse
Affiliation(s)
- Qingyuan Sun
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jinyang Gao
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ran An
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Menggeer Wang
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yanqing Wang
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
5
|
Jiao K, Su P, Li Y. FGFR2 modulates the Akt/Nrf2/ARE signaling pathway to improve angiotensin II-induced hypertension-related endothelial dysfunction. Clin Exp Hypertens 2023; 45:2208777. [PMID: 37154169 DOI: 10.1080/10641963.2023.2208777] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
BACKGROUND Fibroblast growth factor receptor (FGFR)2 expression was decreased in hypertension patients while its role in hypertension was not explored. This experiment aimed to investigate the expression ofFGFR2 in angiotensin II (Ang II)-induced human umbilical vein endothelial cells (HUVECs) and the role of FGFR2 in improving AngII-induced hypertension-related endothelial dysfunction. METHODS AngII-induced HUVECs simulated the hypertension model in vitro. The expression of FGFR2 in Ang II-induced HUVECs and transfected HUVECswas detected by RT-qPCR and western blot. The viability, apoptosis, migration and tube formation ability of Ang II-induced HUVECs were analyzed by Methyl Thiazolyl Tetrazolium (MTT) assay, flow cytometry analysis, wound healing assay and tube formation assay.Detectionof lactate dehydrogenase (LDH), caspase 3, Nitric Oxide (NO) and oxidative stress levels was conducted by assay kits and reactive oxygen species (ROS) level was detected by DCFH-DA assay. The expression of apoptosis-related proteins, protein kinase B(Akt)/nuclear factor E2-related factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathway-related proteins, phospho(p)-endothelial nitric oxide synthase (eNOS) and eNOS was determined by western blot. RESULTS The expression of FGFR2 was decreased in Ang II-induced HUVECs. FGFR2overexpression increased viability, suppressed apoptosis and oxidative stress, and improve endothelial dysfunction of AngII-induced HUVECs through activating the Akt/Nrf2/ARE signaling pathway. MK-2206 (Akt inhibitor) could weaken the effect of FGFR2overexpression to reduce viability, promote apoptosis and oxidative stress, and aggravate endothelial dysfunction of Ang II-inducedHUVECs. CONCLUSION Inconclusion, FGFR2activated the Akt/Nrf2/ARE signaling pathway to improve AngII-induced hypertension-related endothelial dysfunction.
Collapse
Affiliation(s)
- Kun Jiao
- Division 1, Department of Cardiology, Ordos Central Hospital, Inner Mongolia, China
| | - Ping Su
- Division 1, Department of Cardiology, Ordos Central Hospital, Inner Mongolia, China
| | - Yongling Li
- Division 1, Department of Cardiology, Ordos Central Hospital, Inner Mongolia, China
| |
Collapse
|
6
|
Chowdhury FA, Colussi N, Sharma M, Wood KC, Xu JZ, Freeman BA, Schopfer FJ, Straub AC. Fatty acid nitroalkenes - Multi-target agents for the treatment of sickle cell disease. Redox Biol 2023; 68:102941. [PMID: 37907055 PMCID: PMC10632539 DOI: 10.1016/j.redox.2023.102941] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/27/2023] [Accepted: 10/18/2023] [Indexed: 11/02/2023] Open
Abstract
Sickle cell disease (SCD) is a hereditary hematological disease with high morbidity and mortality rates worldwide. Despite being monogenic, SCD patients display a plethora of disease-associated complications including anemia, oxidative stress, sterile inflammation, vaso-occlusive crisis-related pain, and vasculopathy, all of which contribute to multiorgan dysfunction and failure. Over the past decade, numerous small molecule drugs, biologics, and gene-based interventions have been evaluated; however, only four disease-modifying drug therapies are presently FDA approved. Barriers regarding effectiveness, accessibility, affordability, tolerance, and compliance of the current polypharmacy-based disease-management approaches are challenging. As such, there is an unmet pharmacological need for safer, more efficacious, and logistically accessible treatment options for SCD patients. Herein, we evaluate the potential of small molecule nitroalkenes such as nitro-fatty acid (NO2-FA) as a therapy for SCD. These agents are electrophilic and exert anti-inflammatory and tissue repair effects through an ability to transiently post-translationally bind to and modify transcription factors, pro-inflammatory enzymes and cell signaling mediators. Preclinical and clinical studies affirm safety of the drug class and a murine model of SCD reveals protection against inflammation, fibrosis, and vascular dysfunction. Despite protective cardiac, renal, pulmonary, and central nervous system effects of nitroalkenes, they have not previously been considered as therapy for SCD. We highlight the pathways targeted by this drug class, which can potentially prevent the end-organ damage associated with SCD and contrast their prospective therapeutic benefits for SCD as opposed to current polypharmacy approaches.
Collapse
Affiliation(s)
- Fabliha A Chowdhury
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicole Colussi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Malini Sharma
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katherine C Wood
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julia Z Xu
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Division of Hematology and Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Francisco J Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Liver Research Center (PLRC), University of Pittsburgh, Pittsburgh, PA, USA.
| | - Adam C Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Center for Microvascular Research, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Nitro-oleic acid regulates T cell activation through post-translational modification of calcineurin. Proc Natl Acad Sci U S A 2023; 120:e2208924120. [PMID: 36652486 PMCID: PMC9942794 DOI: 10.1073/pnas.2208924120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Nitro-fatty acids (NO2-FAs) are unsaturated fatty acid nitration products that exhibit anti-inflammatory actions in experimental mouse models of autoimmune and allergic diseases. These electrophilic molecules interfere with intracellular signaling pathways by reversible post-translational modification of nucleophilic amino-acid residues. Several regulatory proteins have been identified as targets of NO2-FAs, modifying their activity and promoting gene expression changes that result in anti-inflammatory effects. Herein, we report the effects of nitro-oleic acid (NO2-OA) on pro-inflammatory T cell functions, showing that 9- and 10-NOA, but not their oleic acid precursor, decrease T cell proliferation, expression of activation markers CD25 and CD71 on the plasma membrane, and IL-2, IL-4, and IFN-γ cytokine gene expressions. Moreover, we have found that NO2-OA inhibits the transcriptional activity of nuclear factor of activated T cells (NFAT) and that this inhibition takes place through the regulation of the phosphatase activity of calcineurin (CaN), hindering NFAT dephosphorylation, and nuclear translocation in activated T cells. Finally, using mass spectrometry-based approaches, we have found that NO2-OA nitroalkylates CaNA on four Cys (Cys129, 228, 266, and 372), of which only nitroalkylation on Cys372 was of importance for the regulation of CaN phosphatase activity in cells, disturbing functional CaNA/CaNB heterodimer formation. These results provide evidence for an additional mechanism by which NO2-FAs exert their anti-inflammatory actions, pointing to their potential as therapeutic bioactive lipids for the modulation of harmful T cell-mediated immune responses.
Collapse
|
8
|
Du Y, Zhang H, Nie X, Qi Y, Shi S, Han Y, Zhou W, He C, Wang L. Link between sterile inflammation and cardiovascular diseases: Focus on cGAS-STING pathway in the pathogenesis and therapeutic prospect. Front Cardiovasc Med 2022; 9:965726. [PMID: 36072862 PMCID: PMC9441773 DOI: 10.3389/fcvm.2022.965726] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Sterile inflammation characterized by unresolved chronic inflammation is well established to promote the progression of multiple autoimmune diseases, metabolic disorders, neurodegenerative diseases, and cardiovascular diseases, collectively termed as sterile inflammatory diseases. In recent years, substantial evidence has revealed that the inflammatory response is closely related to cardiovascular diseases. Cyclic guanosine monophosphate–adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway which is activated by cytoplasmic DNA promotes the activation of interferon regulatory factor 3 (IRF3) or nuclear factor-κB (NF-κB), thus leading to upregulation of the levels of inflammatory factors and interferons (IFNs). Therefore, studying the role of inflammation caused by cGAS-STING pathway in cardiovascular diseases could provide a new therapeutic target for cardiovascular diseases. This review focuses on that cGAS-STING-mediated inflammatory response in the progression of cardiovascular diseases and the prospects of cGAS or STING inhibitors for treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yao Du
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hui Zhang
- Department of Stomatology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoyan Nie
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yajun Qi
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Shi Shi
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yingying Han
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenchen Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chaoyong He
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
- *Correspondence: Chaoyong He
| | - Lintao Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
- Lintao Wang
| |
Collapse
|
9
|
Lu Y, Sun Y, Xu K, Saaoud F, Shao Y, Drummer C, Wu S, Hu W, Yu J, Kunapuli SP, Bethea JR, Vazquez-Padron RI, Sun J, Jiang X, Wang H, Yang X. Aorta in Pathologies May Function as an Immune Organ by Upregulating Secretomes for Immune and Vascular Cell Activation, Differentiation and Trans-Differentiation-Early Secretomes may Serve as Drivers for Trained Immunity. Front Immunol 2022; 13:858256. [PMID: 35320939 PMCID: PMC8934864 DOI: 10.3389/fimmu.2022.858256] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/09/2022] [Indexed: 01/09/2023] Open
Abstract
To determine whether aorta becomes immune organ in pathologies, we performed transcriptomic analyses of six types of secretomic genes (SGs) in aorta and vascular cells and made the following findings: 1) 53.7% out of 21,306 human protein genes are classified into six secretomes, namely, canonical, caspase 1, caspase 4, exosome, Weibel-Palade body, and autophagy; 2) Atherosclerosis (AS), chronic kidney disease (CKD) and abdominal aortic aneurysm (AAA) modulate six secretomes in aortas; and Middle East Respiratory Syndrome Coronavirus (MERS-CoV, COVID-19 homologous) infected endothelial cells (ECs) and angiotensin-II (Ang-II) treated vascular smooth muscle cells (VSMCs) modulate six secretomes; 3) AS aortas upregulate T and B cell immune SGs; CKD aortas upregulate SGs for cardiac hypertrophy, and hepatic fibrosis; and AAA aorta upregulate SGs for neuromuscular signaling and protein catabolism; 4) Ang-II induced AAA, canonical, caspase 4, and exosome SGs have two expression peaks of high (day 7)-low (day 14)-high (day 28) patterns; 5) Elastase induced AAA aortas have more inflammatory/immune pathways than that of Ang-II induced AAA aortas; 6) Most disease-upregulated cytokines in aorta may be secreted via canonical and exosome secretomes; 7) Canonical and caspase 1 SGs play roles at early MERS-CoV infected ECs whereas caspase 4 and exosome SGs play roles in late/chronic phases; and the early upregulated canonical and caspase 1 SGs may function as drivers for trained immunity (innate immune memory); 8) Venous ECs from arteriovenous fistula (AVF) upregulate SGs in five secretomes; and 9) Increased some of 101 trained immunity genes and decreased trained tolerance regulator IRG1 participate in upregulations of SGs in atherosclerotic, Ang-II induced AAA and CKD aortas, and MERS-CoV infected ECs, but less in SGs upregulated in AVF ECs. IL-1 family cytokines, HIF1α, SET7 and mTOR, ROS regulators NRF2 and NOX2 partially regulate trained immunity genes; and NRF2 plays roles in downregulating SGs more than that of NOX2 in upregulating SGs. These results provide novel insights on the roles of aorta as immune organ in upregulating secretomes and driving immune and vascular cell differentiations in COVID-19, cardiovascular diseases, inflammations, transplantations, autoimmune diseases and cancers.
Collapse
Affiliation(s)
- Yifan Lu
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Yu Sun
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Keman Xu
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Fatma Saaoud
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Ying Shao
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Charles Drummer
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Sheng Wu
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Wenhui Hu
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Jun Yu
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Satya P. Kunapuli
- Sol Sherry Thrombosis Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - John R. Bethea
- Department of Biology, College of Arts and Sciences, Drexel University, Philadelphia, PA, United States
| | - Roberto I. Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Jianxin Sun
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Hong Wang
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
10
|
Fatty acid nitroalkene reversal of established lung fibrosis. Redox Biol 2021; 50:102226. [PMID: 35150970 PMCID: PMC8844680 DOI: 10.1016/j.redox.2021.102226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue fibrosis occurs in response to dysregulated metabolism, pro-inflammatory signaling and tissue repair reactions. For example, lungs exposed to environmental toxins, cancer therapies, chronic inflammation and other stimuli manifest a phenotypic shift to activated myofibroblasts and progressive and often irreversible lung tissue scarring. There are no therapies that stop or reverse fibrosis. The 2 FDA-approved anti-fibrotic drugs at best only slow the progression of fibrosis in humans. The present study was designed to test whether a small molecule electrophilic nitroalkene, nitro-oleic acid (NO2-OA), could reverse established pulmonary fibrosis induced by the intratracheal administration of bleomycin in C57BL/6 mice. After 14 d of bleomycin-induced fibrosis development in vivo, lungs were removed, sectioned and precision-cut lung slices (PCLS) from control and bleomycin-treated mice were cultured ex vivo for 4 d with either vehicle or NO2-OA (5 μM). Biochemical and morphological analyses showed that over a 4 d time frame, NO2-OA significantly inhibited pro-inflammatory mediator and growth factor expression and reversed key indices of fibrosis (hydroxyproline, collagen 1A1 and 3A1, fibronectin-1). Quantitative image analysis of PCLS immunohistology reinforced these observations, revealing that NO2-OA suppressed additional hallmarks of the fibrotic response, including alveolar epithelial cell loss, myofibroblast differentiation and proliferation, collagen and α-smooth muscle actin expression. NO2-OA also accelerated collagen degradation by resident macrophages. These effects occurred in the absence of the recognized NO2-OA modulation of circulating and migrating immune cell activation. Thus, small molecule nitroalkenes may be useful agents for reversing pathogenic fibrosis of lung and other organs. Small molecule electrophiles, pleiotropic anti-inflammatory and anti-fibrotic drugs. NO2-OA inhibits activated myofibroblasts, induces dedifferentiation to fibroblasts. NO2-OA activates extracellular matrix degradation by macrophages. NO2-OA promotes proliferation of alveolar type 1 and 2 epithelial cells. NO2-OA reverses established lung fibrosis in murine lung slices.
Collapse
|
11
|
Pereckova J, Pekarova M, Szamecova N, Hoferova Z, Kamarytova K, Falk M, Perecko T. Nitro-Oleic Acid Inhibits Stemness Maintenance and Enhances Neural Differentiation of Mouse Embryonic Stem Cells via STAT3 Signaling. Int J Mol Sci 2021; 22:ijms22189981. [PMID: 34576143 PMCID: PMC8468660 DOI: 10.3390/ijms22189981] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/19/2021] [Accepted: 09/12/2021] [Indexed: 12/15/2022] Open
Abstract
Nitro-oleic acid (NO2-OA), pluripotent cell-signaling mediator, was recently described as a modulator of the signal transducer and activator of transcription 3 (STAT3) activity. In our study, we discovered new aspects of NO2-OA involvement in the regulation of stem cell pluripotency and differentiation. Murine embryonic stem cells (mESC) or mESC-derived embryoid bodies (EBs) were exposed to NO2-OA or oleic acid (OA) for selected time periods. Our results showed that NO2-OA but not OA caused the loss of pluripotency of mESC cultivated in leukemia inhibitory factor (LIF) rich medium via the decrease of pluripotency markers (NANOG, sex-determining region Y-box 1 transcription factor (SOX2), and octamer-binding transcription factor 4 (OCT4)). The effects of NO2-OA on mESC correlated with reduced phosphorylation of STAT3. Subsequent differentiation led to an increase of the ectodermal marker orthodenticle homolog 2 (Otx2). Similarly, treatment of mESC-derived EBs by NO2-OA resulted in the up-regulation of both neural markers Nestin and β-Tubulin class III (Tubb3). Interestingly, the expression of cardiac-specific genes and beating of EBs were significantly decreased. In conclusion, NO2-OA is able to modulate pluripotency of mESC via the regulation of STAT3 phosphorylation. Further, it attenuates cardiac differentiation on the one hand, and on the other hand, it directs mESC into neural fate.
Collapse
Affiliation(s)
- Jana Pereckova
- Institute of Biophysics of the Czech Academy of Sciences, Department of Cell Biology and Radiobiology, Kralovopolska 135, 612 65 Brno, Czech Republic; (M.P.); (N.S.); (Z.H.); (K.K.); (M.F.); (T.P.)
- Correspondence:
| | - Michaela Pekarova
- Institute of Biophysics of the Czech Academy of Sciences, Department of Cell Biology and Radiobiology, Kralovopolska 135, 612 65 Brno, Czech Republic; (M.P.); (N.S.); (Z.H.); (K.K.); (M.F.); (T.P.)
| | - Nikoletta Szamecova
- Institute of Biophysics of the Czech Academy of Sciences, Department of Cell Biology and Radiobiology, Kralovopolska 135, 612 65 Brno, Czech Republic; (M.P.); (N.S.); (Z.H.); (K.K.); (M.F.); (T.P.)
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Zuzana Hoferova
- Institute of Biophysics of the Czech Academy of Sciences, Department of Cell Biology and Radiobiology, Kralovopolska 135, 612 65 Brno, Czech Republic; (M.P.); (N.S.); (Z.H.); (K.K.); (M.F.); (T.P.)
| | - Kristyna Kamarytova
- Institute of Biophysics of the Czech Academy of Sciences, Department of Cell Biology and Radiobiology, Kralovopolska 135, 612 65 Brno, Czech Republic; (M.P.); (N.S.); (Z.H.); (K.K.); (M.F.); (T.P.)
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Martin Falk
- Institute of Biophysics of the Czech Academy of Sciences, Department of Cell Biology and Radiobiology, Kralovopolska 135, 612 65 Brno, Czech Republic; (M.P.); (N.S.); (Z.H.); (K.K.); (M.F.); (T.P.)
| | - Tomas Perecko
- Institute of Biophysics of the Czech Academy of Sciences, Department of Cell Biology and Radiobiology, Kralovopolska 135, 612 65 Brno, Czech Republic; (M.P.); (N.S.); (Z.H.); (K.K.); (M.F.); (T.P.)
| |
Collapse
|
12
|
Zhou C, Su M, Sun P, Tang X, Yin KJ. Nitro-oleic acid-mediated blood-brain barrier protection reduces ischemic brain injury. Exp Neurol 2021; 346:113861. [PMID: 34499902 DOI: 10.1016/j.expneurol.2021.113861] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/28/2021] [Accepted: 09/05/2021] [Indexed: 10/20/2022]
Abstract
Nitro-oleic acid (OA-NO2), a nitroalkene formed in nitric oxide-dependent oxidative reactions, has been found in human plasma and is thought to regulate pathophysiological functions. Recently, accumulating evidence suggests that OA-NO2 may function as an anti-inflammatory mediator, and ameliorate the progression of diabetes and cardiovascular diseases. However, the role of OA-NO2 in ischemic brain injury remains unexplored. In this study, C57BL/6 mice were subjected to 1 h transient middle cerebral artery occlusion (MCAO) and followed by 1- 7 days of reperfusion. These mice were treated with vehicle, OA, or OA-NO2 (10 mg/kg) via tail vein injection at 2 h after the onset of MCAO. Our results show that intravenous administration of OA-NO2 led to reduced BBB leakage in ischemic brains, reduced brain infarct, and improved sensorimotor functions in response to ischemic insults when compared to OA and vehicle controls. Also, OA-NO2 significantly reduced BBB leakage-triggered infiltration of neutrophils and macrophages in the ischemic brains. Moreover, OA-NO2 treatment reduced the M1-type microglia and increased M2-type microglia. Mechanistically, OA-NO2 alleviated the decline of mRNA and protein level of major endothelial TJs including ZO-1 in stroke mice. Treatment of OA-NO2 also significantly inhibited stroke-induced inflammatory mediators, iNOS, E-selectin, P-selectin, and ICAM1, in mouse brains. In conclusion, OA-NO2 preserves BBB integrity and confers neurovascular protection in ischemic brain damage. OA-NO2-mediated brain protection may help us to develop a novel therapeutic strategy for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Chao Zhou
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Moxi Su
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ping Sun
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xuelian Tang
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ke-Jie Yin
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
13
|
Hidalgo MA, Carretta MD, Burgos RA. Long Chain Fatty Acids as Modulators of Immune Cells Function: Contribution of FFA1 and FFA4 Receptors. Front Physiol 2021; 12:668330. [PMID: 34276398 PMCID: PMC8280355 DOI: 10.3389/fphys.2021.668330] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022] Open
Abstract
Long-chain fatty acids are molecules that act as metabolic intermediates and constituents of membranes; however, their novel role as signaling molecules in immune function has also been demonstrated. The presence of free fatty acid (FFA) receptors on immune cells has contributed to the understanding of this new role of long-chain fatty acids (LCFAs) in immune function, showing their role as anti-inflammatory or pro-inflammatory molecules and elucidating their intracellular mechanisms. The FFA1 and FFA4 receptors, also known as GPR40 and GPR120, respectively, have been described in macrophages and neutrophils, two key cells mediating innate immune response. Ligands of the FFA1 and FFA4 receptors induce the release of a myriad of cytokines through well-defined intracellular signaling pathways. In this review, we discuss the cellular responses and intracellular mechanisms activated by LCFAs, such as oleic acid, linoleic acid, palmitic acid, docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA), in T-cells, macrophages, and neutrophils, as well as the role of the FFA1 and FFA4 receptors in immune cells.
Collapse
Affiliation(s)
- Maria A Hidalgo
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| | - Maria D Carretta
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| | - Rafael A Burgos
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
14
|
Lin K, Luo W, Yan J, Shen S, Shen Q, Wang J, Guan X, Wu G, Huang W, Liang G. TLR2 regulates angiotensin II-induced vascular remodeling and EndMT through NF-κB signaling. Aging (Albany NY) 2020; 13:2553-2574. [PMID: 33318302 PMCID: PMC7880316 DOI: 10.18632/aging.202290] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022]
Abstract
Excessive vascular remodeling has been shown in hypertensive patients. In experimental models of hypertensive vascular injury, such as angiotensin II (Ang II) challenged mice, toll like receptor 2 (TLR2) initiates inflammatory responses. More recently, studies have reported atypical endothelial to mesenchymal transition (EndMT) in vascular injuries and inflammatory conditions. Here, we aimed to investigate whether TLR2 mediates Ang II-induced vascular inflammation and initiates EndMT. In a mouse model of angiotensin II-induced hypertension, we show that aortas exhibit increased medial thickening, fibrosis, and features of EndMT. These alterations were not observed in TLR2 knockout mice in response to Ang II. TLR2 silencing in cultured endothelial cells confirmed the essential role of TLR2 in Ang II-induced inflammatory factor induction, and EndMT-associated phenotypic change. Mechanistically, we found Ang II activates nuclear factor-κB signaling, inducing pro-inflammatory cytokine production, and mediates EndMT in both cultured endothelial cells and in mice. These studies illustrate a novel role of TLR2 in regulating Ang II-induced deleterious vascular remodeling through the induction of EndMT. The studies also suggest that TLR2 may be targeted to alleviate hypertension-associated vascular injury.
Collapse
Affiliation(s)
- Ke Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.,Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jueqian Yan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Siyuan Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.,Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Qirui Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jun Wang
- Department of Cardiology, Wenzhou Central Hospital and Affiliated Dingli Clinical Institute, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Xinfu Guan
- Affiliated Cangnan Hospital, Wenzhou Medical University, Cangnan 325800, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.,Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.,Affiliated Cangnan Hospital, Wenzhou Medical University, Cangnan 325800, Zhejiang, China
| |
Collapse
|
15
|
O’Brien J, Wendell SG. Electrophile Modulation of Inflammation: A Two-Hit Approach. Metabolites 2020; 10:metabo10110453. [PMID: 33182676 PMCID: PMC7696920 DOI: 10.3390/metabo10110453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022] Open
Abstract
Electrophilic small molecules have gained significant attention over the last decade in the field of covalent drug discovery. Long recognized as mediators of the inflammatory process, recent evidence suggests that electrophiles may modulate the immune response through the regulation of metabolic networks. These molecules function as pleiotropic signaling mediators capable of reversibly reacting with nucleophilic biomolecules, most notably at reactive cysteines. More specifically, electrophiles target critical cysteines in redox regulatory proteins to activate protective pathways such as the nuclear factor erythroid 2-related factor 2-Kelch-like ECH-associated protein 1 (Nrf2-Keap1) antioxidant signaling pathway while also inhibiting Nuclear Factor κB (NF-κB). During inflammatory states, reactive species broadly alter cell signaling through the oxidation of lipids, amino acids, and nucleic acids, effectively propagating the inflammatory sequence. Subsequent changes in metabolic signaling inform immune cell maturation and effector function. Therapeutic strategies targeting inflammatory pathologies leverage electrophilic drug compounds, in part, because of their documented effect on the redox balance of the cell. With mounting evidence demonstrating the link between redox signaling and metabolism, electrophiles represent ideal therapeutic candidates for the treatment of inflammatory conditions. Through their pleiotropic signaling activity, electrophiles may be used strategically to both directly and indirectly target immune cell metabolism.
Collapse
|
16
|
Mata-Pérez C, Padilla MN, Sánchez-Calvo B, Begara-Morales JC, Valderrama R, Chaki M, Aranda-Caño L, Moreno-González D, Molina-Díaz A, Barroso JB. Endogenous Biosynthesis of S-Nitrosoglutathione From Nitro-Fatty Acids in Plants. FRONTIERS IN PLANT SCIENCE 2020; 11:962. [PMID: 32714353 PMCID: PMC7340149 DOI: 10.3389/fpls.2020.00962] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/11/2020] [Indexed: 05/05/2023]
Abstract
Nitro-fatty acids (NO2-FAs) are novel molecules resulting from the interaction of unsaturated fatty acids and nitric oxide (NO) or NO-related molecules. In plants, it has recently been described that NO2-FAs trigger an antioxidant and a defence response against stressful situations. Among the properties of NO2-FAs highlight the ability to release NO therefore modulating specific protein targets through post-translational modifications (NO-PTMs). Thus, based on the capacity of NO2-FAs to act as physiological NO donors and using high-accuracy mass-spectrometric approaches, herein, we show that endogenous nitro-linolenic acid (NO2-Ln) can modulate S-nitrosoglutathione (GSNO) biosynthesis in Arabidopsis. The incubation of NO2-Ln with GSH was analyzed by LC-MS/MS and the in vitro synthesis of GSNO was noted. The in vivo confirmation of this behavior was carried out by incubating Arabidopsis plants with 15N-labeled NO2-Ln throughout the roots, and 15N-labeled GSNO (GS15NO) was detected in the leaves. With the aim to go in depth in the relation of NO2-FA and GSNO in plants, Arabidopsis alkenal reductase mutants (aer mutants) which modulate NO2-FAs levels were used. Our results constitute the first evidence of the modulation of a key NO biological reservoir in plants (GSNO) by these novel NO2-FAs, increasing knowledge about S-nitrosothiols and GSNO-signaling pathways in plants.
Collapse
Affiliation(s)
- Capilla Mata-Pérez
- Group of Biochemistry and Cell Signalling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - María N. Padilla
- Group of Biochemistry and Cell Signalling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - Beatriz Sánchez-Calvo
- Group of Biochemistry and Cell Signalling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - Juan C. Begara-Morales
- Group of Biochemistry and Cell Signalling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - Raquel Valderrama
- Group of Biochemistry and Cell Signalling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - Mounira Chaki
- Group of Biochemistry and Cell Signalling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - Lorena Aranda-Caño
- Group of Biochemistry and Cell Signalling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - David Moreno-González
- Analytical Chemistry Research Group, Department of Physical and Analytical Chemistry, University of Jaén, Jaén, Spain
| | - Antonio Molina-Díaz
- Analytical Chemistry Research Group, Department of Physical and Analytical Chemistry, University of Jaén, Jaén, Spain
| | - Juan B. Barroso
- Group of Biochemistry and Cell Signalling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- *Correspondence: Juan B. Barroso,
| |
Collapse
|
17
|
Tang H, Mao J, Ye X, Zhang F, Kerr WG, Zheng T, Zhu Z. SHIP-1, a target of miR-155, regulates endothelial cell responses in lung fibrosis. FASEB J 2019; 34:2011-2023. [PMID: 31907997 DOI: 10.1096/fj.201902063r] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/22/2019] [Accepted: 11/01/2019] [Indexed: 12/12/2022]
Abstract
Src Homology 2-containing Inositol Phosphatase-1 (SHIP-1) is a target of miR-155, a pro-inflammatory factor. Deletion of the SHIP-1 gene in mice caused spontaneous lung inflammation and fibrosis. However, the role and function of endothelial miR-155 and SHIP-1 in lung fibrosis remain unknown. Using whole-body miR-155 knockout mice and endothelial cell-specific conditional miR-155 (VEC-Cre-miR-155 or VEC-miR-155) or SHIP-1 (VEC-SHIP-1) knockout mice, we assessed endothelial-mesenchymal transition (EndoMT) and fibrotic responses in bleomycin (BLM) induced lung fibrosis models. Primary mouse lung endothelial cells (MLEC) and human umbilical vein endothelial cells (HUVEC) with SHIP-1 knockdown were analyzed in TGF-β1 or BLM, respectively, induced fibrotic responses. Fibrosis and EndoMT were significantly reduced in miR-155KO mice and changes in EndoMT markers in MLEC after TGF-β1 stimulation confirmed the in vivo findings. Furthermore, lung fibrosis and EndoMT responses were reduced in VEC-miR-155 mice but significantly enhanced in VEC-SHIP-1 mice after BLM challenge. SHIP-1 knockdown in HUVEC cells resulted in enhanced EndoMT induced by BLM. Meanwhile, these changes involved the PI3K/AKT, JAK/STAT3, and SMAD/STAT signaling pathways. These studies demonstrate that endothelial miR-155 plays an important role in fibrotic responses in the lung through EndoMT. Endothelial SHIP-1 is essential in controlling fibrotic responses and SHIP-1 is a target of miR-155. Endothelial cells are an integral part in lung fibrosis.
Collapse
Affiliation(s)
- Haiying Tang
- Section of Allergy and Clinical Immunology, Yale University School of Medicine, New Haven, CT, USA.,Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jingwei Mao
- Section of Allergy and Clinical Immunology, Yale University School of Medicine, New Haven, CT, USA.,Department of Gastroenterology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xujun Ye
- Section of Allergy and Clinical Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - Fengrui Zhang
- Section of Allergy and Clinical Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - William G Kerr
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Tao Zheng
- Section of Allergy and Clinical Immunology, Yale University School of Medicine, New Haven, CT, USA.,Department of Molecular Microbiology and Immunology, Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Zhou Zhu
- Section of Allergy and Clinical Immunology, Yale University School of Medicine, New Haven, CT, USA.,Department of Molecular Microbiology and Immunology, Brown University Warren Alpert Medical School, Providence, RI, USA
| |
Collapse
|
18
|
Su W, Wang H, Feng Z, Sun J. Nitro-oleic acid inhibits the high glucose-induced epithelial-mesenchymal transition in peritoneal mesothelial cells and attenuates peritoneal fibrosis. Am J Physiol Renal Physiol 2019; 318:F457-F467. [PMID: 31760768 DOI: 10.1152/ajprenal.00425.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
As an electrophilic nitroalkene fatty acid, nitro-oleic acid (OA-NO2) exerts multiple biological effects that contribute to anti-inflammation, anti-oxidative stress, and antiapoptosis. However, little is known about the role of OA-NO2 in peritoneal fibrosis. Thus, in the present study, we examined the effects of OA-NO2 on the high glucose (HG)-induced epithelial-mesenchymal transition (EMT) in human peritoneal mesothelial cells (HPMCs) and evaluated the morphological and immunohistochemical changes in a rat model of peritoneal dialysis-related peritoneal fibrosis. In in vitro experiments, we found that HG reduced the expression level of E-cadherin and increased Snail, N-cadherin, and α-smooth muscle actin expression levels in HPMCs. The above-mentioned changes were attenuated by pretreatment with OA-NO2. Additionally, OA-NO2 also inhibited HG-induced activation of the transforming growth factor-β1/Smad signaling pathway and NF-κB signaling pathway. Meanwhile, OA-NO2 inhibited HG-induced phosphorylation of Erk and JNK. The results from the in vivo experiments showed that OA-NO2 notably relieved peritoneal fibrosis by decreasing the thickness of the peritoneum; it also inhibited expression of transforming growth factor-β1, α-smooth muscle actin, N-cadherin, and vimentin and enhanced expression of E-cadherin in the peritoneum. Collectively, these results suggest that OA-NO2 inhibits the HG-induced epithelial-mesenchymal transition in HPMCs and attenuates peritoneal dialysis-related peritoneal fibrosis.
Collapse
Affiliation(s)
- Wenyan Su
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shangdong, China
| | - Haiping Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shangdong, China
| | - ZiYan Feng
- Department of Dialysis, JuanCheng People's Hospital, Heze, Shangdong, China
| | - Jing Sun
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shangdong, China
| |
Collapse
|
19
|
You S, Qian J, Wu G, Qian Y, Wang Z, Chen T, Wang J, Huang W, Liang G. Schizandrin B attenuates angiotensin II induced endothelial to mesenchymal transition in vascular endothelium by suppressing NF-κB activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 62:152955. [PMID: 31146168 DOI: 10.1016/j.phymed.2019.152955] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Angiotensin II (Ang II)-induced chronic inflammation and oxidative stress often leads to irreversible vascular injury, in which the endothelial to mesenchymal transition (EndMT) in the endothelial layers are involved. Schisandrin B (Sch B), a natural product isolated from traditional Schisandra chinensis, has been reported to exert vascular protective properties with unclear mechanism. HYPOTHESIS/PURPOSE This study investigated the protective effects and mechanism of Sch B against Ang II-induced vascular injury. METHODS C57BL/6 mice were subcutaneous injected of Ang II for 4 weeks to induce irreversible vascular injury. In vitro, Ang II-induced HUVECs injury was used to study the underlying mechanism. The markers of EndMT, inflammation and oxidative stress were studied both in vitro and in vivo. RESULTS Pre-administration of Sch B effectively attenuated phenotypes of vascular EndMT and fibrosis in Ang II-treated animals, accompanied with decreased inflammatory cytokine and ROS. The in vitro data from HUVECs suggest that Sch B directly targets NF-κB activation to suppress Ang II-induced EndMT and vascular injury. The activation of EndMT in the presence of Ang II is regulated by the NF-κB, a common player in inflammation and oxidative stress. Ang II-induced inflammation and oxidative stress also contributed to vascular EndMT development and Sch B inhibited inflammation/ROS-mediated EndMT by suppressing NF-κB. CONCLUSION EndMT contributes to vascular injury in Ang II-treated mice, and it can be prevented via suppressing NF-κB activation by Sch B treatment. These results also imply that NF-κB might be a promising target to attenuate vascular remodeling induced by inflammation and oxidative stress through an EndMT mechanism.
Collapse
Affiliation(s)
- Shengban You
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianchang Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuanyuan Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhengxian Wang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Taiwei Chen
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingying Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
20
|
Piera-Velazquez S, Jimenez SA. Endothelial to Mesenchymal Transition: Role in Physiology and in the Pathogenesis of Human Diseases. Physiol Rev 2019; 99:1281-1324. [PMID: 30864875 DOI: 10.1152/physrev.00021.2018] [Citation(s) in RCA: 385] [Impact Index Per Article: 64.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Numerous studies have demonstrated that endothelial cells are capable of undergoing endothelial to mesenchymal transition (EndMT), a newly recognized type of cellular transdifferentiation. EndMT is a complex biological process in which endothelial cells adopt a mesenchymal phenotype displaying typical mesenchymal cell morphology and functions, including the acquisition of cellular motility and contractile properties. Endothelial cells undergoing EndMT lose the expression of endothelial cell-specific proteins such as CD31/platelet-endothelial cell adhesion molecule, von Willebrand factor, and vascular-endothelial cadherin and initiate the expression of mesenchymal cell-specific genes and the production of their encoded proteins including α-smooth muscle actin, extra domain A fibronectin, N-cadherin, vimentin, fibroblast specific protein-1, also known as S100A4 protein, and fibrillar type I and type III collagens. Transforming growth factor-β1 is considered the main EndMT inducer. However, EndMT involves numerous molecular and signaling pathways that are triggered and modulated by multiple and often redundant mechanisms depending on the specific cellular context and on the physiological or pathological status of the cells. EndMT participates in highly important embryonic development processes, as well as in the pathogenesis of numerous genetically determined and acquired human diseases including malignant, vascular, inflammatory, and fibrotic disorders. Despite intensive investigation, many aspects of EndMT remain to be elucidated. The identification of molecules and regulatory pathways involved in EndMT and the discovery of specific EndMT inhibitors should provide novel therapeutic approaches for various human disorders mediated by EndMT.
Collapse
Affiliation(s)
- Sonsoles Piera-Velazquez
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Aranda-Caño L, Sánchez-Calvo B, Begara-Morales JC, Chaki M, Mata-Pérez C, Padilla MN, Valderrama R, Barroso JB. Post-Translational Modification of Proteins Mediated by Nitro-Fatty Acids in Plants: Nitroalkylation. PLANTS 2019; 8:plants8040082. [PMID: 30934982 PMCID: PMC6524050 DOI: 10.3390/plants8040082] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/28/2022]
Abstract
Nitrate fatty acids (NO₂-FAs) are considered reactive lipid species derived from the non-enzymatic oxidation of polyunsaturated fatty acids by nitric oxide (NO) and related species. Nitrate fatty acids are powerful biological electrophiles which can react with biological nucleophiles such as glutathione and certain protein⁻amino acid residues. The adduction of NO₂-FAs to protein targets generates a reversible post-translational modification called nitroalkylation. In different animal and human systems, NO₂-FAs, such as nitro-oleic acid (NO₂-OA) and conjugated nitro-linoleic acid (NO₂-cLA), have cytoprotective and anti-inflammatory influences in a broad spectrum of pathologies by modulating various intracellular pathways. However, little knowledge on these molecules in the plant kingdom exists. The presence of NO₂-OA and NO₂-cLA in olives and extra-virgin olive oil and nitro-linolenic acid (NO₂-Ln) in Arabidopsis thaliana has recently been detected. Specifically, NO₂-Ln acts as a signaling molecule during seed and plant progression and beneath abiotic stress events. It can also release NO and modulate the expression of genes associated with antioxidant responses. Nevertheless, the repercussions of nitroalkylation on plant proteins are still poorly known. In this review, we demonstrate the existence of endogenous nitroalkylation and its effect on the in vitro activity of the antioxidant protein ascorbate peroxidase.
Collapse
Affiliation(s)
- Lorena Aranda-Caño
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University Campus Las Lagunillas, University of Jaén, E-23071 Jaén, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Rom O, Khoo NKH, Chen YE, Villacorta L. Inflammatory signaling and metabolic regulation by nitro-fatty acids. Nitric Oxide 2018; 78:S1089-8603(17)30329-4. [PMID: 29578057 PMCID: PMC6151155 DOI: 10.1016/j.niox.2018.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 02/06/2023]
Abstract
The addition of nitrogen dioxide (NO2) to the double bond of unsaturated fatty acids yields an array of electrophilic nitro-fatty acids (NO2-FA) with unique biochemical and signaling properties. During the last decade, NO2-FA have been shown to exert a protective role in various inflammatory and metabolic disorders. NO2-FA exert their biological effects primarily by regulating two central physiological adaptive responses: the canonical inflammatory signaling and metabolic pathways. In this mini-review, we summarize current knowledge on the regulatory role of NO2-FA in the inflammatory and metabolic response via regulation of nuclear factor kappa B (NF-κB) and peroxisome proliferator-activated receptor γ (PPARγ), master regulators of inflammation and metabolism. Moreover, the engagement of novel signaling and metabolic pathways influenced by NO2-FA, beyond NF-κB and PPAR signaling, is discussed herein.
Collapse
Affiliation(s)
- Oren Rom
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, USA
| | - Nicholas K H Khoo
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, USA
| | - Y Eugene Chen
- Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, USA
| | - Luis Villacorta
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, USA.
| |
Collapse
|
23
|
Yi F, Hao Y, Chong X, Zhong W. Overexpression of microRNA-506-3p aggravates the injury of vascular endothelial cells in patients with hypertension by downregulating Beclin1 expression. Exp Ther Med 2018; 15:2844-2850. [PMID: 29456688 PMCID: PMC5795718 DOI: 10.3892/etm.2018.5733] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 06/29/2017] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to measure the expression of microRNA (miRNA)-506-3p in the peripheral blood of patients with hypertension and to determine the biological functions and mechanisms of action of miR-506-3p. A total of 61 patients with primary hypertension were included in the present study. Peripheral blood was collected from all patients, as well as 31 healthy subjects who were included in a control group. The expression of miR-506-3p in peripheral blood was determined by reverse transcription-quantitative polymerase chain reaction. Human umbilical vein endothelial cells (HUVECs) were transfected with miR-506-3p mimics or miR-506-3p inhibitor. The proliferation and migration of HUVECs were determined using cell-counting kit 8 and Transwell assays, respectively. The cell cycle and apoptosis of HUVECs were detected by flow cytometry. The expression of Beclin1 (BECN1) protein, a potential target of miR-506-3p, was measured using western blotting. A dual-luciferase reporter assay was performed to determine the interaction between BECN1 and miR-506-3p. It was demonstrated that miR-506-3p expression in the peripheral blood of patients with patients was upregulated and dependent on the severity of hypertension. miR-506-3p overexpression inhibited the proliferation and migration of HUVECs. In addition, miR-506-3p inhibited the transition from the G1 phase to the S-phase in HUVECs. Overexpression of miR-506-3p promoted the apoptosis of HUVECs. Notably, miR-506-3p downregulated the expression of BECN1 by directly binding to its 3′-untranslated region. The present study demonstrated that miR-506-3p expression is elevated in the peripheral blood of patients with hypertension and is associated with the severity of hypertension. By downregulating BECN1 expression, miR-506-3p aggravates injury in vascular endothelial cells by inhibiting the proliferation and migration of HUVECs, as well as promoting their apoptosis.
Collapse
Affiliation(s)
- Fanfan Yi
- Department of Emergency, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277101, P.R. China
| | - Yugui Hao
- Department of Emergency, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277101, P.R. China
| | - Xiaoyi Chong
- Department of Clinical Medicine, Medical College of Qinghai University, Xining, Qinghai 810000, P.R. China
| | - Wei Zhong
- Cadre Ward, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277101, P.R. China
| |
Collapse
|
24
|
Mata-Pérez C, Sánchez-Calvo B, Padilla MN, Begara-Morales JC, Valderrama R, Corpas FJ, Barroso JB. Nitro-fatty acids in plant signaling: New key mediators of nitric oxide metabolism. Redox Biol 2017; 11:554-561. [PMID: 28104576 PMCID: PMC5241575 DOI: 10.1016/j.redox.2017.01.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 12/30/2016] [Accepted: 01/03/2017] [Indexed: 01/21/2023] Open
Abstract
Recent studies in animal systems have shown that NO can interact with fatty acids to generate nitro-fatty acids (NO2-FAs). They are the product of the reaction between reactive nitrogen species and unsaturated fatty acids, and are considered novel mediators of cell signaling based mainly on a proven anti-inflammatory response. Although these signaling mediators have been described widely in animal systems, NO2-FAs have scarcely been studied in plants. Preliminary data have revealed the endogenous presence of free and protein-adducted NO2-FAs in extra-virgin olive oil (EVOO), which appear to be contributing to the cardiovascular benefits associated with the Mediterranean diet. Importantly, new findings have displayed the endogenous occurrence of nitro-linolenic acid (NO2-Ln) in the model plant Arabidopsis thaliana and the modulation of NO2-Ln levels throughout this plant's development. Furthermore, a transcriptomic analysis by RNA-seq technology established a clear signaling role for this molecule, demonstrating that NO2-Ln was involved in plant-defense response against different abiotic-stress conditions, mainly by inducing the chaperone network and supporting a conserved mechanism of action in both animal and plant defense processes. Thus, NO2-Ln levels significantly rose under several abiotic-stress conditions, highlighting the strong signaling role of these molecules in the plant-protection mechanism. Finally, the potential of NO2-Ln as a NO donor has recently been described both in vitro and in vivo. Jointly, this ability gives NO2-Ln the potential to act as a signaling molecule by the direct release of NO, due to its capacity to induce different changes mediated by NO or NO-related molecules such as nitration and S-nitrosylation, or by the electrophilic capacity of these molecules through a nitroalkylation mechanism. Here, we describe the current state of the art regarding the advances performed in the field of NO2-FAs in plants and their implication in plant physiology.
Collapse
Affiliation(s)
- Capilla Mata-Pérez
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, Campus Universitario "Las Lagunillas" s/n, University of Jaén, E-23071 Jaén, Spain
| | - Beatriz Sánchez-Calvo
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, Campus Universitario "Las Lagunillas" s/n, University of Jaén, E-23071 Jaén, Spain
| | - María N Padilla
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, Campus Universitario "Las Lagunillas" s/n, University of Jaén, E-23071 Jaén, Spain
| | - Juan C Begara-Morales
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, Campus Universitario "Las Lagunillas" s/n, University of Jaén, E-23071 Jaén, Spain
| | - Raquel Valderrama
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, Campus Universitario "Las Lagunillas" s/n, University of Jaén, E-23071 Jaén, Spain
| | - Francisco J Corpas
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cellular and Molecular Biology of Plants, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Juan B Barroso
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, Campus Universitario "Las Lagunillas" s/n, University of Jaén, E-23071 Jaén, Spain.
| |
Collapse
|
25
|
Koudelka A, Ambrozova G, Klinke A, Fidlerova T, Martiskova H, Kuchta R, Rudolph TK, Kadlec J, Kuchtova Z, Woodcock SR, Freeman BA, Kubala L, Pekarova M. Nitro-Oleic Acid Prevents Hypoxia- and Asymmetric Dimethylarginine-Induced Pulmonary Endothelial Dysfunction. Cardiovasc Drugs Ther 2017; 30:579-586. [PMID: 27858190 DOI: 10.1007/s10557-016-6700-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE Pulmonary hypertension (PH) represents a serious health complication accompanied with hypoxic conditions, elevated levels of asymmetric dimethylarginine (ADMA), and overall dysfunction of pulmonary vascular endothelium. Since the prevention strategies for treatment of PH remain largely unknown, our study aimed to explore the effect of nitro-oleic acid (OA-NO2), an exemplary nitro-fatty acid (NO2-FA), in human pulmonary artery endothelial cells (HPAEC) under the influence of hypoxia or ADMA. METHODS HPAEC were treated with OA-NO2 in the absence or presence of hypoxia and ADMA. The production of nitric oxide (NO) and interleukin-6 (IL-6) was monitored using the Griess method and ELISA, respectively. The expression or activation of different proteins (signal transducer and activator of transcription 3, STAT3; hypoxia inducible factor 1α, HIF-1α; endothelial nitric oxide synthase, eNOS; intercellular adhesion molecule-1, ICAM-1) was assessed by the Western blot technique. RESULTS We discovered that OA-NO2 prevents development of endothelial dysfunction induced by either hypoxia or ADMA. OA-NO2 preserves normal cellular functions in HPAEC by increasing NO production and eNOS expression. Additionally, OA-NO2 inhibits IL-6 production as well as ICAM-1 expression, elevated by hypoxia and ADMA. Importantly, the effect of OA-NO2 is accompanied by prevention of STAT3 activation and HIF-1α stabilization. CONCLUSION In summary, OA-NO2 eliminates the manifestation of hypoxia- and ADMA-mediated endothelial dysfunction in HPAEC via the STAT3/HIF-1α cascade. Importantly, our study is bringing a new perspective on molecular mechanisms of NO2-FAs action in pulmonary endothelial dysfunction, which represents a causal link in progression of PH. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Adolf Koudelka
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic
| | - Gabriela Ambrozova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic
| | - Anna Klinke
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.,Department of Experimental Cardiology, University Hospital of Cologne, Kerpener Str. 62, 50924, Cologne, Germany
| | - Tana Fidlerova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic.,Faculty of Science, Institute of Experimental Biology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Hana Martiskova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.,Faculty of Science, Institute of Experimental Biology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Radek Kuchta
- Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3058/10, 616 00, Brno, Czech Republic
| | - Tanja K Rudolph
- Department of Experimental Cardiology, University Hospital of Cologne, Kerpener Str. 62, 50924, Cologne, Germany
| | - Jaroslav Kadlec
- Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3058/10, 616 00, Brno, Czech Republic
| | - Zdenka Kuchtova
- Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3058/10, 616 00, Brno, Czech Republic
| | - Steven R Woodcock
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
| | - Lukas Kubala
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic
| | - Michaela Pekarova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 612 65, Brno, Czech Republic. .,International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.
| |
Collapse
|
26
|
Verescakova H, Ambrozova G, Kubala L, Perecko T, Koudelka A, Vasicek O, Rudolph TK, Klinke A, Woodcock SR, Freeman BA, Pekarova M. Nitro-oleic acid regulates growth factor-induced differentiation of bone marrow-derived macrophages. Free Radic Biol Med 2017; 104:10-19. [PMID: 28063941 PMCID: PMC5329068 DOI: 10.1016/j.freeradbiomed.2017.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/20/2016] [Accepted: 01/03/2017] [Indexed: 01/05/2023]
Abstract
Many diseases accompanied by chronic inflammation are connected with dysregulated activation of macrophage subpopulations. Recently, we reported that nitro-fatty acids (NO2-FAs), products of metabolic and inflammatory reactions of nitric oxide and nitrite, modulate macrophage and other immune cell functions. Bone marrow cell suspensions were isolated from mice and supplemented with macrophage colony-stimulating factor (M-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF) in combination with NO2-OA for different times. RAW 264.7 macrophages were used for short-term (1-5min) experiments. We discovered that NO2-OA reduces cell numbers, cell colony formation, and proliferation of macrophages differentiated with colony-stimulating factors (CSFs), all in the absence of toxicity. In a case of GM-CSF-induced bone marrow-derived macrophages (BMMs), NO2-OA acts via downregulation of signal transducer and activator of transcription 5 and extracellular signal-regulated kinase (ERK) activation. In the case of M-CSF-induced BMMs, NO2-OA decreases activation of M-CSFR and activation of related PI3K and ERK. Additionally, NO2-OA also attenuates activation of BMMs. In aggregate, we demonstrate that NO2-OA regulates the process of macrophage differentiation and that NO2-FAs represent a promising therapeutic tool in the treatment of inflammatory pathologies linked with increased accumulation of macrophages in inflamed tissues.
Collapse
Affiliation(s)
- Hana Verescakova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia
| | - Gabriela Ambrozova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia
| | - Lukas Kubala
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia
| | - Tomas Perecko
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia
| | - Adolf Koudelka
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia; Department of Animal Physiology and Immunology, Masaryk University, Brno, Czechia
| | - Ondrej Vasicek
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia
| | - Tanja K Rudolph
- Heart Centre, University Hospital of Cologne, Cologne, Germany
| | - Anna Klinke
- International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia; Heart Centre, University Hospital of Cologne, Cologne, Germany
| | - Steven R Woodcock
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michaela Pekarova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia.
| |
Collapse
|
27
|
Shang J, Zhang Y, Jiang Y, Li Z, Duan Y, Wang L, Xiao J, Zhao Z. NOD2 promotes endothelial-to-mesenchymal transition of glomerular endothelial cells via MEK/ERK signaling pathway in diabetic nephropathy. Biochem Biophys Res Commun 2017; 484:435-441. [PMID: 28137583 DOI: 10.1016/j.bbrc.2017.01.155] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 01/26/2017] [Indexed: 12/22/2022]
Abstract
Endothelial-to-mesenchymal transition (EndMT) of glomerular vascular endothelial cells (GEnCs) is now considered to play a critical role in diabetic nephropathy (DN). NOD2 is newly discovered to be closely related to DN renal injury. However, the relationship between NOD2 and EndMT of GEnCs has never been reported. In the present study, we found that NOD2 over-expression was positively correlated with the severity of DN injury in human renal biopsy samples. Immunohistochemical staining of DN renal slices showed gradual absence of endothelial character and gain of mesenchymal character, both of which were associated with NOD2 over-expression. In high glucose stimulated GEnCs, NOD2 was increased. What's more, over-expression and activation of NOD2 could both promote EndMT of GEnCs. On the other hand, silencing of NOD2 markedly attenuated EndMT induced by high glucose. Mechanically, we further found that MEK/ERK signaling pathway was involved in NOD2-regulated EndMT. Collectively, our results indicate that NOD2 has a regulatory role in EndMT via activation of MEK/ERK in high glucose-treated GEnCs. Targeting this pathway is a promising strategy for intervention of DN endothelial dysfunction.
Collapse
Affiliation(s)
- Jin Shang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ya Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yumin Jiang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenzhen Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiqi Duan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Luyao Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Xiao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhanzheng Zhao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
28
|
Electrophilic Nitro-Fatty Acids: Nitric Oxide and Nitrite-Derived Metabolic and Inflammatory Signaling Mediators. Nitric Oxide 2017. [DOI: 10.1016/b978-0-12-804273-1.00016-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|